1
|
Singh R, Panghal A, Jadhav K, Thakur A, Verma RK, Singh C, Goyal M, Kumar J, Namdeo AG. Recent Advances in Targeting Transition Metals (Copper, Iron, and Zinc) in Alzheimer's Disease. Mol Neurobiol 2024; 61:10916-10940. [PMID: 38809370 DOI: 10.1007/s12035-024-04256-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Changes in the transition metal homeostasis in the brain are closely linked with Alzheimer's disease (AD), including intraneuronal iron accumulation and extracellular copper and zinc pooling in the amyloid plague. The brain copper, zinc, and iron surplus are commonly acknowledged characteristics of AD, despite disagreements among some. This has led to the theory that oxidative stress resulting from abnormal homeostasis of these transition metals may be a causative explanation behind AD. In the nervous system, the interaction of metals with proteins appears to be an essential variable in the development or suppression of neurodegeneration. Chelation treatment may be an option for treating neurodegeneration induced by transition metal ion dyshomeostasis. Some clinicians even recommend using chelating agents as an adjunct therapy for AD. The current review also looks at the therapeutic strategies that have been attempted, primarily with metal-chelating drugs. Metal buildup in the nervous system, as reported in the AD, could be the result of compensatory mechanisms designed to improve metal availability for physiological functions.
Collapse
Affiliation(s)
- Raghuraj Singh
- Pharmaceutical Nanotechnology Lab, Institutes of Nano Science and Technology (INST), Sector 81. Mohali, Punjab, 140306, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Archna Panghal
- Department of Pharmacology and Toxicology, Facility for Risk Assessment and Intervention Studies, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, India
| | - Krishna Jadhav
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Ashima Thakur
- Faculty of Pharmaceutical Sciences, ICFAI University, Baddi, Distt. Solan, Himachal Pradesh, 174103, India
| | - Rahul Kumar Verma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Charan Singh
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India
| | - Manoj Goyal
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India
| | - Jayant Kumar
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India.
| | - Ajay G Namdeo
- Department of Pharmaceutical Sciences Hemwati, Nandan Bahuguna Garhwal University (A Central University), Srinagar, Dist. Garhwal (Uttarakhand), 246174, India
| |
Collapse
|
2
|
Hossain MS, Haque MA, Park IS. Novel role of curcumin as inhibitor of β-amyloid-induced lamin fragmentation. Histochem Cell Biol 2024; 163:2. [PMID: 39542878 DOI: 10.1007/s00418-024-02331-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 11/17/2024]
Abstract
Oligomer amyloid beta 42 (Aβ) is considered the key pathogenic molecule in Alzheimer disease (AD) and causes specific lamin fragmentation. Curcumin has been recognized for its protective effects against Aβ-induced toxicity in AD, though its underlying mechanism remains unclear. In this study, the inhibitory mechanism of curcumin against Aβ-induced lamin fragmentation and cell death was investigated. Human neuroblastoma cells were used to examine Aβ-induced lamin fragmentation and lamin deformation by immunoblotting and confocal microscopy, while cell viability was measured using MTT and alamarBlue assay. Caspase and cathepsin L activity were assessed by spectrofluorometry, and Aβ aggregation was evaluated by ThT assay. Our results demonstrated that curcumin inhibited Aβ aggregation, reducing intracellular Aβ uptake by 45% compared to Aβ-treated cells. Curcumin also inhibited the Aβ-induced intracellular calcium rise, subsequently leading to a onefold reduction in cathepsin L activity. This reduction in cathepsin L activity by curcumin blocked the Aβ-induced lamin fragmentation. Collectively, these findings suggest that curcumin inhibits Aβ-induced cell death by preventing Aβ entry and lamin cleavage, providing potential new insights for AD treatment.
Collapse
Affiliation(s)
- Md Selim Hossain
- Department of Biomedical Sciences, Chosun University, Gwangju, 61452, Republic of Korea.
- Department of Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| | - Md Aminul Haque
- Department of Biomedical Sciences, Chosun University, Gwangju, 61452, Republic of Korea
- School of Pharmacy, BRAC University, Dhaka, Bangladesh
- Rufaida BioMeds, Dhaka, Bangladesh
| | - Il-Seon Park
- Department of Biomedical Sciences, Chosun University, Gwangju, 61452, Republic of Korea.
- Department of Cellular and Molecular Medicine, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
3
|
Duranti E, Villa C. Insights into Dysregulated Neurological Biomarkers in Cancer. Cancers (Basel) 2024; 16:2680. [PMID: 39123408 PMCID: PMC11312413 DOI: 10.3390/cancers16152680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The link between neurodegenerative diseases (NDs) and cancer has generated greater interest in biomedical research, with decades of global studies investigating neurodegenerative biomarkers in cancer to better understand possible connections. Tau, amyloid-β, α-synuclein, SOD1, TDP-43, and other proteins associated with nervous system diseases have also been identified in various types of solid and malignant tumors, suggesting a potential overlap in pathological processes. In this review, we aim to provide an overview of current evidence on the role of these proteins in cancer, specifically examining their effects on cell proliferation, apoptosis, chemoresistance, and tumor progression. Additionally, we discuss the diagnostic and therapeutic implications of this interconnection, emphasizing the importance of further research to completely comprehend the clinical implications of these proteins in tumors. Finally, we explore the challenges and opportunities in targeting these proteins for the development of new targeted anticancer therapies, providing insight into how to integrate knowledge of NDs in oncology research.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
4
|
Midorikawa R, Wakazono Y, Takamiya K. Aβ peptide enhances GluA1 internalization via lipid rafts in Alzheimer's-related hippocampal LTP dysfunction. J Cell Sci 2024; 137:jcs261281. [PMID: 38668720 DOI: 10.1242/jcs.261281] [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: 04/26/2023] [Accepted: 03/08/2024] [Indexed: 05/01/2024] Open
Abstract
Amyloid β (Aβ) is a central contributor to neuronal damage and cognitive impairment in Alzheimer's disease (AD). Aβ disrupts AMPA receptor-mediated synaptic plasticity, a key factor in early AD progression. Numerous studies propose that Aβ oligomers hinder synaptic plasticity, particularly long-term potentiation (LTP), by disrupting GluA1 (encoded by GRIA1) function, although the precise mechanism remains unclear. In this study, we demonstrate that Aβ mediates the accumulation of GM1 ganglioside in lipid raft domains of cultured cells, and GluA1 exhibits preferential localization in lipid rafts via direct binding to GM1. Aβ enhances the raft localization of GluA1 by increasing GM1 in these areas. Additionally, chemical LTP stimulation induces lipid raft-dependent GluA1 internalization in Aβ-treated neurons, resulting in reduced cell surface and postsynaptic expression of GluA1. Consistent with this, disrupting lipid rafts and GluA1 localization in rafts rescues Aβ-mediated suppression of hippocampal LTP. These findings unveil a novel functional deficit in GluA1 trafficking induced by Aβ, providing new insights into the mechanism underlying AD-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Ryosuke Midorikawa
- Department of Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yoshihiko Wakazono
- Department of Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
- Laboratory of Biophysical Research, Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Kogo Takamiya
- Department of Neuroscience, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
- Laboratory of Biophysical Research, Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan
| |
Collapse
|
5
|
James E, Vielle A, Cusato K, Li H, Lee B, Parween S, Howell A, Johnson NR, Chial HJ, Potter H, Vergara MN. Human iPSC-derived retinal organoids develop robust Alzheimer's disease neuropathology. Front Cell Neurosci 2024; 18:1340448. [PMID: 38323188 PMCID: PMC10844524 DOI: 10.3389/fncel.2024.1340448] [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: 11/18/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024] Open
Abstract
Alzheimer's disease (AD), characterized by memory loss and cognitive decline, affects nearly 50 million people worldwide. Amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) of phosphorylated Tau protein (pTau) are key histopathological features of the disease in the brain, and recent advances have also identified AD histopathology in the retina. Thus, the retina represents a central nervous system (CNS) tissue highly amenable to non-invasive diagnostic imaging that shows promise as a biomarker for early AD. Given the devastating effects of AD on patients, their families, and society, new treatment modalities that can significantly alter the disease course are urgently needed. In this study, we have developed and characterized a novel human retinal organoid (RO) model derived from induced pluripotent stem cells (iPSCs) from patients with familial AD due to mutations in the amyloid precursor protein gene (APP). Using immunofluorescence and histological staining, we evaluated the cellular composition and AD histopathological features of AD-ROs compared to control ROs from healthy individuals. We found that AD-ROs largely resemble their healthy control counterparts in cellular composition but display increased levels of Aβ and pTau. We also present proof of principle of an assay to quantify amyloid levels in whole ROs. This in vitro model of the human AD retina constitutes a new tool for drug screening, biomarker discovery, and pathophysiological studies.
Collapse
Affiliation(s)
- Ethan James
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Anne Vielle
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Karen Cusato
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Helen Li
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Byoungin Lee
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Shama Parween
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Anna Howell
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Noah R. Johnson
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Heidi J. Chial
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Huntington Potter
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - M. Natalia Vergara
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
6
|
Madar P, Nagalapur P, Chaudhari S, Sharma D, Koparde A, Buchade R, Kshirsagar S, Uttekar P, Jadhav S, Chaudhari P. The Unveiling of Therapeutic Targets for Alzheimer's Disease: An Integrative Review. Curr Top Med Chem 2024; 24:850-868. [PMID: 38424435 DOI: 10.2174/0115680266282492240220101049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
Alzheimer's disease (AD) is characterized by a complex pathological landscape, necessitating a comprehensive treatment approach. This concise review paper delves into the idea of addressing multiple mechanisms in AD, summarizing the latest research findings on pathogenesis, risk factors, diagnostics, and therapeutic strategies. The etiology of AD is multifaceted, involving genetic, environmental, and lifestyle factors. The primary feature is the accumulation of amyloid-- beta and tau proteins, leading to neuroinflammation, synaptic dysfunction, oxidative stress, and neuronal loss. Conventional single-target therapies have shown limited effectiveness, prompting a shift toward simultaneously addressing multiple disease-related processes. Recent advancements in AD research underscore the potential of multifaceted therapies. This review explores strategies targeting both tau aggregation and amyloid-beta, along with interventions to alleviate neuroinflammation, enhance synaptic function, and reduce oxidative stress. In conclusion, the review emphasizes the growing importance of addressing various pathways in AD treatment. A holistic approach that targets different aspects of the disease holds promise for developing effective treatments and improving the quality of life for Alzheimer's patients and their caregivers.
Collapse
Affiliation(s)
- Pratiksha Madar
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| | - Pooja Nagalapur
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| | - Somdatta Chaudhari
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| | - Devesh Sharma
- Department of Biotechnology, National JALMA Institute for Leprosy & Other Mycobacterial Diseases, Agra, India
| | - Akshada Koparde
- Department of Pharmaceutical Chemistry, Krishna Foundation's Jaywant Institute of Pharmacy, Malkapur, Karad, India
| | - Rahul Buchade
- Department of Pharmaceutical Chemistry, Indira College of Pharmacy, Tathwade, Pune, India
| | - Sandip Kshirsagar
- Department of Pharmaceutical Chemistry, Dr. D Y Patil College of Pharmacy, Pune, India
| | - Pravin Uttekar
- Department of Pharmacuetics, Savitribai Phule Pune University, Pune, India
| | - Shailaja Jadhav
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| | - Praveen Chaudhari
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
7
|
Chen J, Tang F, Li H, Wu X, Yang Y, Liu Z, Huang X, Wang J, Zheng R, Wang L, Liu H, Xu J, Wang P, Liu F. Mycobacterium tuberculosis suppresses APLP2 expression to enhance its survival in macrophage. Int Immunopharmacol 2023; 124:111058. [PMID: 37844466 DOI: 10.1016/j.intimp.2023.111058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
Mycobacterium tuberculosis (M.tb), the most successful pathogen responsible for approximately 1.6 million deaths in 2021, employs various strategies to evade host antibacterial defenses, including mechanisms to counteract nitric oxide (NO) and certain cytokines. While Amyloid β (A4) precursor-like protein 2 (Aplp2) has been implicated in various physiological and pathological processes, its role in tuberculosis (TB) pathogenesis remains largely uncharted. This study unveils a significant reduction in Aplp2 levels in TB patients, M.tb-infected macrophages, and mice. Intriguingly, Aplp2 mutation or knockdown results in diminished macrophage-mediated killing of M.tb, accompanied by decreased inducible nitric oxide synthase (iNOS) expression and reduced cytokine production, notably interleukin-1β (Il-1β). Notably, Aplp2 mutant mice exhibit heightened susceptibility to mycobacterial infection, evident through aggravated histopathological damage and increased lung bacterial loads, in contrast to Mycobacterium bovis BCG-infected wild-type (WT) mice. Mechanistically, the cleaved product of APLP2, AICD2, generated by γ-secretase, translocates to the nucleus, where it interacts with p65, culminating in enhanced the nuclear factor κB (NF-κB) transcriptional activity. This interaction triggers the upregulation of Il-1β and iNOS expression. Collectively, our findings illuminate Aplp2's pivotal role in safeguarding against mycobacterial infections by promoting M.tb clearance through NO- or IL-1β-mediated bactericidal effects. Therefore, we unveil a novel immune evasion strategy employed by M.tb, which could potentially serve as a target for innovative TB interventions.
Collapse
Affiliation(s)
- Jianxia Chen
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Fen Tang
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Haohao Li
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xiangyang Wu
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Yong Yang
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Zhonghua Liu
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xiaochen Huang
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Jie Wang
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Ruijuan Zheng
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Lin Wang
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Haipeng Liu
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Junfang Xu
- Clinical and Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Peng Wang
- Department of TB, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Feng Liu
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai JiaoTong University, Shanghai Sixth People's Hospital Affiliated to JiaoTong University Medical School, Shanghai 200233, China.
| |
Collapse
|
8
|
Basli A, Bounaas J. Pathophysiological mechanism and natural preventive and therapeutic strategies of Alzheimer's disease. Nutr Health 2023; 29:403-413. [PMID: 36377316 DOI: 10.1177/02601060221137104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) is characterized by the presence of two types of protein deposits in the brain, amyloid plaques and neurofibrillary tangles. The first one are dense deposits of beta amyloid protein, the second one are dense deposits of the protein tau. These proteins are present in all of our brains, but in AD they act unusually, leading to neuronal degeneration. This review will provide an overview of the AD, including the role of amyloid beta and tau, and mechanisms that lead to the formation of plaques and tangles. The review will also cover the existing researches that have focused on the inhibition of amyloid beta formation, cholinesterase, tau hyperphosphorylation, the pathogenic mechanisms of apoE4, and GSK-3 as a solution that could be used to slow or prevent the disease.
Collapse
Affiliation(s)
- Abdelkader Basli
- Laboratory of Interaction Research, Biodiversity, Ecosystems and Biotechnology, Faculty of Sciences, University of Skikda, Skikda, Algeria
| | - Jihane Bounaas
- Laboratory of Interaction Research, Biodiversity, Ecosystems and Biotechnology, Faculty of Sciences, University of Skikda, Skikda, Algeria
| |
Collapse
|
9
|
Malter JS. Pin1 and Alzheimer's disease. Transl Res 2023; 254:24-33. [PMID: 36162703 PMCID: PMC10111655 DOI: 10.1016/j.trsl.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/29/2022] [Accepted: 09/19/2022] [Indexed: 10/14/2022]
Abstract
Alzheimer's disease (AD) is an immense and growing public health crisis. Despite over 100 years of investigation, the etiology remains elusive and therapy ineffective. Despite current gaps in knowledge, recent studies have identified dysfunction or loss-of-function of Pin1, a unique cis-trans peptidyl prolyl isomerase, as an important step in AD pathogenesis. Here I review the functionality of Pin1 and its role in neurodegeneration.
Collapse
Affiliation(s)
- James S Malter
- Department of Pathology, UT Southwestern Medical Center, 5333 Harry Hines Blvd, Dallas, TX 75390.
| |
Collapse
|
10
|
Yanagida K, Maruyama R, Tagami S, Kudo T, Okochi M, Fukumori A. APLP2 is predominantly cleaved by β-secretase and γ-secretase in the human brain. Psychogeriatrics 2023; 23:311-318. [PMID: 36691315 DOI: 10.1111/psyg.12933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/25/2022] [Accepted: 01/03/2023] [Indexed: 01/25/2023]
Abstract
BACKGROUND Amyloid-β peptide is well-known as a pathogen of Alzheimer's disease, but its precursor, amyloid-beta precursor protein (APP), remains unexplained 30 years after its discovery. APP has two homologues called amyloid precursor-like protein 1 (APLP1) and amyloid precursor-like protein 2 (APLP2), and shares a similar structural organisation with them and has partially overlapping functions. APP family proteins are essential for survival, shown by the crossbreeding analysis of knockout mice of APP family molecules, including APLP1 and APLP2. APLP2 is known to play the most important role among them, but the molecular metabolism of APLP2 is only partially understood. Here, we analysed ectodomain shedding and γ-secretase cleavage of APLP2 by molecular biological and biochemical techniques. METHOD We analysed the culture supernatant of HEK293 cells overexpressing APLP2 and human cerebrospinal fluid. For the analysis of secreted APLP2 fragments, we raised the OA603 antibody that reacts with the juxtamembrane domain of APLP2. Substrate cleavage sites were identified by matrix assisted laser desorption/ionisation mass spectrometry. RESULTS By overexpressing in HEK293 cells, APLP2 undergoes ectodomain shedding at three sites in the extracellular region by α- and β-secretase-like activity and then is intramembranously cleaved at three sites by γ-secretase. In particular, in shedding, α-secretase-like activity was dominant in HEK cells. Surprisingly, in human cerebrospinal fluid, APLP2-derived metabolic fragments were mainly cleaved by β-secretase-like activity, not by α-secretase-like activity. Because APP is also mainly cleaved by beta-site amyloid precursor protein cleaving enzyme 1 in neurons and APLP1 is expressed exclusively in neurons, these findings suggest that APP family proteins may play a common role via β-secretase-like cleavage in the central nerve system. CONCLUSIONS Thus, these findings may contribute to a better understanding of the role of APP family proteins in Alzheimer's disease.
Collapse
Affiliation(s)
- Kanta Yanagida
- Department of Mental Health Promotion, Osaka University Graduate School of Medicine, Toyonaka, Japan.,Department of Pharmacotherapeutics II, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Japan.,Neuropsychiatry, Department of Integrated Medicine, Division of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Riki Maruyama
- Department of Mental Health Promotion, Osaka University Graduate School of Medicine, Toyonaka, Japan.,Department of Pharmacotherapeutics II, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Shinji Tagami
- Neuropsychiatry, Department of Integrated Medicine, Division of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takashi Kudo
- Department of Mental Health Promotion, Osaka University Graduate School of Medicine, Toyonaka, Japan
| | - Masayasu Okochi
- Neuropsychiatry, Department of Integrated Medicine, Division of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Akio Fukumori
- Department of Mental Health Promotion, Osaka University Graduate School of Medicine, Toyonaka, Japan.,Department of Pharmacotherapeutics II, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Japan.,Neuropsychiatry, Department of Integrated Medicine, Division of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, Obu, Japan
| |
Collapse
|
11
|
TRPV1 Modulator Ameliorates Alzheimer-Like Amyloid- β Neuropathology via Akt/Gsk3 β-Mediated Nrf2 Activation in the Neuro-2a/APP Cell Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1544244. [PMID: 36065437 PMCID: PMC9440841 DOI: 10.1155/2022/1544244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 08/01/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disorder for which there is no effective therapeutic strategy. PcActx peptide from the transcriptome of zoantharian Palythoa caribaeorum has recently been identified and verified as a novel antagonist of transient receptor potential cation channel subfamily V member 1 (TRPV1). In the present study, we further investigated the neuroprotective potential of PcActx peptide and its underlying mechanism of action, in an N2a/APP cell model of AD. Both Western blot and RT-PCR analysis revealed that PcActx peptide markedly inhibited the production of amyloid-related proteins and the expression of BACE1, PSEN1, and PSEN2. Moreover, PcActx peptide notably attenuated the capsaicin-stimulated calcium response and prevented the phosphorylation of CaMKII and CaMKIV (calcium-mediated proteins) in N2a/APP cells. Further investigation indicated that PcActx peptide significantly suppressed ROS generation through Nrf2 activation, followed by enhanced NQO1 and HO-1 levels. In addition, PcActx peptide remarkably improved Akt phosphorylation at Ser 473 (active) and Gsk3β phosphorylation at Ser 9 (inactive), while pharmacological inhibition of the Akt/Gsk3β pathway significantly attenuated PcActx-induced Nrf2 activation and amyloid downregulation. In conclusion, PcActx peptide functions as a TRPV1 modulator of intercellular calcium homeostasis, prevents AD-like amyloid neuropathology via Akt/Gsk3β-mediated Nrf2 activation, and shows promise as an alternative therapeutic agent for AD.
Collapse
|
12
|
Pfundstein G, Nikonenko AG, Sytnyk V. Amyloid precursor protein (APP) and amyloid β (Aβ) interact with cell adhesion molecules: Implications in Alzheimer’s disease and normal physiology. Front Cell Dev Biol 2022; 10:969547. [PMID: 35959488 PMCID: PMC9360506 DOI: 10.3389/fcell.2022.969547] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is an incurable neurodegenerative disorder in which dysfunction and loss of synapses and neurons lead to cognitive impairment and death. Accumulation and aggregation of neurotoxic amyloid-β (Aβ) peptides generated via amyloidogenic processing of amyloid precursor protein (APP) is considered to play a central role in the disease etiology. APP interacts with cell adhesion molecules, which influence the normal physiological functions of APP, its amyloidogenic and non-amyloidogenic processing, and formation of Aβ aggregates. These cell surface glycoproteins also mediate attachment of Aβ to the neuronal cell surface and induce intracellular signaling contributing to Aβ toxicity. In this review, we discuss the current knowledge surrounding the interactions of cell adhesion molecules with APP and Aβ and analyze the evidence of the critical role these proteins play in regulating the processing and physiological function of APP as well as Aβ toxicity. This is a necessary piece of the complex AD puzzle, which we should understand in order to develop safe and effective therapeutic interventions for AD.
Collapse
Affiliation(s)
- Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | | | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
- *Correspondence: Vladimir Sytnyk,
| |
Collapse
|
13
|
Zhang H, Ben Zablah Y, Liu A, Lee D, Zhang H, Meng Y, Zhou C, Liu X, Wang Y, Jia Z. Overexpression of LIMK1 in hippocampal excitatory neurons improves synaptic plasticity and social recognition memory in APP/PS1 mice. Mol Brain 2021; 14:121. [PMID: 34315506 PMCID: PMC8314529 DOI: 10.1186/s13041-021-00833-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/16/2021] [Indexed: 01/22/2023] Open
Abstract
Accumulating evidence indicates that the actin regulator cofilin is overactivated in Alzheimer's Disease (AD), but whether this abnormality contributes to synaptic and cognitive impairments in AD is unclear. In addition, the brain region and cell types involved remain unknown. In this study, we specifically manipulate LIMK1, the key protein kinase that phosphorylates and inactivates cofilin, in the hippocampus of APP/PS1 transgenic mice. Using local injections of the AAV virus containing LIMK1 under the control of the CaMKIIα promoter, we show that expression of LIMK1 in hippocampal excitatory neurons increases cofilin phosphorylation (i.e., decreases cofilin activity), rescues impairments in long-term potentiation, and improves social memory in APP/PS1 mice. Our results suggest that deficits in LIMK1/cofilin signaling in the hippocampal excitatory neurons contribute to AD pathology and that manipulations of LIMK1/cofilin activity provide a potential therapeutic strategy to treat AD.
Collapse
Affiliation(s)
- Haiwang Zhang
- Guizhou Medical University, Guiyang, 550000, Guizhou, China.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, M5S 1A8, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Youssif Ben Zablah
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, M5S 1A8, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - An Liu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Dongju Lee
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, M5S 1A8, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Haorui Zhang
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, M5S 1A8, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Yanghong Meng
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, M5S 1A8, Canada.,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Changxi Zhou
- Department of Geriatrics, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, China
| | - Xingde Liu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, Guizhou, China
| | - Yiming Wang
- Department of Psychiatry, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China. .,Mental Health Education and Counseling Center for College Students, Guizhou Medical University, Guiyang, 550004, Guizhou, China.
| | - Zhengping Jia
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, M5S 1A8, Canada. .,Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
14
|
Molecular Characteristics of Amyloid Precursor Protein (APP) and Its Effects in Cancer. Int J Mol Sci 2021; 22:ijms22094999. [PMID: 34066808 PMCID: PMC8125876 DOI: 10.3390/ijms22094999] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022] Open
Abstract
Amyloid precursor protein (APP) is a type 1 transmembrane glycoprotein, and its homologs amyloid precursor-like protein 1 (APLP1) and amyloid precursor-like protein 2 (APLP2) are highly conserved in mammals. APP and APLP are known to be intimately involved in the pathogenesis and progression of Alzheimer's disease and to play important roles in neuronal homeostasis and development and neural transmission. APP and APLP are also expressed in non-neuronal tissues and are overexpressed in cancer cells. Furthermore, research indicates they are involved in several cancers. In this review, we examine the biological characteristics of APP-related family members and their roles in cancer.
Collapse
|
15
|
APLP2 gene polymorphisms are associated with high TC and LDL-C levels in Chinese population in Xinjiang, China. Biosci Rep 2021; 40:225897. [PMID: 32716039 PMCID: PMC7403944 DOI: 10.1042/bsr20200357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 01/16/2023] Open
Abstract
Hyperlipidemia is one of the main risk factors for coronary artery disease (CAD). In the present study, we aimed to explore whether the single-nucleotide polymorphisms (SNPs) in amyloid precursor-like protein (APLP) 2 (APLP2) gene were associated with high lipid levels in Chinese population in Xinjiang, China. We recruited 1738 subjects (1187 men, 551 women) from the First Affiliated Hospital of Xinjiang Medical University, and genotyped three SNPs (rs2054247, rs3740881 and rs747180) of APLP2 gene in all subjects by using the improved multiplex ligation detection reaction (iMLDR) method. Our study revealed that the rs2054247 SNP was associated with serum total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) levels, and high-density lipoprotein cholesterol (HDL-C) in additive model (all P<0.05). The rs747180 SNP was associated with serum TC and LDL-C levels in additive model (all P<0.05). Our study revealed that both rs2054247 and rs747180 SNPs of the APLP2 gene were associated with high TC and LDL-C levels in Chinese subjects in Xinjiang.
Collapse
|
16
|
Islam MI, Hossain MS, Park IS. Differential involvement of caspase-6 in amyloid-β-induced fragmentation of lamin A and B. Biochem Biophys Rep 2020; 24:100839. [PMID: 33145443 PMCID: PMC7591731 DOI: 10.1016/j.bbrep.2020.100839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/12/2020] [Accepted: 10/22/2020] [Indexed: 01/01/2023] Open
Abstract
Amyloid-β (Aβ), a peptide implicated in Alzheimer's disease, was shown to cause specific fragmentation of lamin proteins, which was mediated by an unidentified protease named nuclear scaffold protease (NSP) independently of caspase-6. Because caspase-6 is responsible for the fragmentation process in many other damage-induced apoptosis, here we further investigated possible involvement of caspase-6 in Aβ-induced lamin fragmentation under various conditions. We found that lamin A fragment generated by NSP (named fragment b) disappeared in cells incubated with Aβ42 for prolonged periods and this product was preserved by a caspase-6 inhibitor. Furthermore, caspase-6 could remove fragment b in nuclei isolated from Aβ42-treated cells (ANU). Lamin B in ANU was fragmented by caspase-6 only after treatment with an alkaline phosphatase. The caspase-mediated fragmentation of lamin B was also achieved with nuclei isolated from cells incubated with Aβ42 plus a Cdk5 inhibitor. The results indicate that Aβ42 induces NSP-mediated fragmentation of lamin A and the following removal process of fragment b by caspase-6 and an Aβ-induced phosphorylation prevents the fragmentation of lamin B by caspase-6. The pathway leading to lamin protein fragmentation in this investigation appears to be specific for Aβ and thus the data will provide novel insights into the toxicity of the peptide. Aβ42 induces nuclear scaffold protease (NSP)-mediated fragmentation of lamin A. The produced fragment of lamin A is subsequently removed by caspase-6. Aβ42 also induced NSP-mediated lamin B fragmentation. Caspase-6-mediated fragmentation of lamin B protein is absent. The absence appears to be due to phosphorylation of lamin B.
Collapse
Affiliation(s)
| | | | - Il-Seon Park
- Department of Medical Sciences, Republic of Korea.,Department of Cellular and Molecular Medicine, Chosun University, Gwanju, 501-759, Republic of Korea
| |
Collapse
|
17
|
Ahmed GAR, Khalil SKH, Hotaby WE, Abbas L, Farrag ARH, Aal WEA, Sherif HHA, Abdel-Rahman EA, Saber SH, Hassan M, Hassan MH, Balgoon M, Qusti S, Kotb M, Ali SS. ATR-IR and EPR spectroscopy for following the membrane restoration of isolated cortical synaptosomes in aluminium-induced Alzheimer's disease - Like rat model. Chem Phys Lipids 2020; 231:104931. [PMID: 32619464 DOI: 10.1016/j.chemphyslip.2020.104931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/22/2020] [Accepted: 06/01/2020] [Indexed: 01/03/2023]
Abstract
Synaptosomal membrane peroxidation and alteration in its biophysical properties are associated with Aluminium (Al) toxicity that may lead to cognitive dysfunction and Alzheimer's disease (AD) like pathogenesis. Here we investigated the therapeutic potential of Lepedium sativum (LS) as a natural anti-inflammatory, antioxidant and as acetyl cholinesterase inhibitor in treating Al induced AD-like in rat model. We utilized ATR-IR spectroscopy to follow the restoration in the damaged membrane structure of isolated rat cortical synaptosomes and its biophysical properties, electron paramagnetic resonance (EPR) spin trapping to follow NADPH oxidase activity (NOX), and EPR spin labelling in response to LS treatment after Al intoxication. We measured the concentration of Ca2+ ions in rat cortical tissue by inductively coupled plasma (ICP), the brain atrophy/curing and hydrocephalus by magnetic resonance imaging (MRI) besides light microscope histopathology. Our results revealed significant increase in synaptosomal membrane rgidification, order, lipid packing, reactive oxygen species (ROS) production and Ca2+ ion concentration as a result of Al intoxication. The dramatic increase in Ca2+ ion concentration detected in AD group associated with the increase in synaptic membrane polarity and EPR-detected order S-parameter suggest that release of synaptic vesicles into synaptic cleft might be hindered. LS treatment reversed these changes in synaptic membranes, and rescued an observed deficit in the exploratory behaviour of AD group. Our results also strongly suggest that the synaptosomal membrane phospholipids that underwent free radical attacks mediated by AlCl3, due to greater NOX activity, was prevented in the LS group. The results of ATR-IR and EPR spectroscopic techniques recommend LS as a promising therapeutic agent against synaptic membrane alterations opening a new window for AD drug developers.
Collapse
Affiliation(s)
- Gehan A-R Ahmed
- Spectroscopy Dept., Physics Division, National Research Centre, 33 El Bohouth St., (former El Tahrir St.), Dokki, Giza, P.O. 12622, Egypt; Biochemistry Dept., Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Safaa K H Khalil
- Spectroscopy Dept., Physics Division, National Research Centre, 33 El Bohouth St., (former El Tahrir St.), Dokki, Giza, P.O. 12622, Egypt
| | - W El Hotaby
- Spectroscopy Dept., Physics Division, National Research Centre, 33 El Bohouth St., (former El Tahrir St.), Dokki, Giza, P.O. 12622, Egypt
| | - Lamyaa Abbas
- Spectroscopy Dept., Physics Division, National Research Centre, 33 El Bohouth St., (former El Tahrir St.), Dokki, Giza, P.O. 12622, Egypt
| | | | - Wafaa E Abdel Aal
- Pathology Dept., Medical Research Div., National Research Centre, Giza, Egypt
| | - Hadeer H A Sherif
- Spectroscopy Dept., Physics Division, National Research Centre, 33 El Bohouth St., (former El Tahrir St.), Dokki, Giza, P.O. 12622, Egypt
| | - Engy A Abdel-Rahman
- Center for Aging and Associated Diseases, Helmy Institute of Medical Sciences, Zewail City of Science and Technology, Giza, Egypt; Pharmacology Department, Faculty of Medicine, Assuit University, Assuit, Egypt; Children's Cancer Hospital 57357, Cair, Egypt
| | - Saber H Saber
- Center for Aging and Associated Diseases, Helmy Institute of Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Mahmoud Hassan
- Center for Aging and Associated Diseases, Helmy Institute of Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
| | - Mohamed H Hassan
- Centre for Materials Science, Zewail City of Science and Technology, Giza, Egypt
| | - Maha Balgoon
- Biochemistry Dept., Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Safaa Qusti
- Biochemistry Dept., Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mamdooh Kotb
- Department of Radiology, King Abdulaziz University, Hospital, Jeddah, Saudi Arabia
| | - Sameh S Ali
- Center for Aging and Associated Diseases, Helmy Institute of Medical Sciences, Zewail City of Science and Technology, Giza, Egypt; Children's Cancer Hospital 57357, Cair, Egypt.
| |
Collapse
|
18
|
Alzheimer's disease; a review of the pathophysiological basis and therapeutic interventions. Life Sci 2020; 256:117996. [PMID: 32585249 DOI: 10.1016/j.lfs.2020.117996] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/14/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and is identified as the most common cause for dementia. Despite huge global economic burden and the impact on the close family of the patients, there is no definitive cure and thus, improved treatment methods are of need. While memory and cognition are severely affected in AD, exact etiology is yet unknown. The β-Amyloid plaque formation and aggregation hypothesis is among the well-known hypotheses used to explain disease pathogenesis. Currently there are five Food and Drug Administration (FDA) approved drugs as treatment options. All these drugs are used for symptomatic treatment of AD. Thus, disease modifying therapies which can directly address the pathological changes in AD, are needed. Such therapies could be designed based on inhibiting key steps of pathogenesis. Currently there are novel AD drug candidates with various therapeutic mechanisms, undergoing different stages of drug development. Extensive research is being done globally to broaden understanding of the exact mechanisms involved in AD and to develop therapeutic agents that can successfully hinder the occurrence and progression of the disease. In this review, a comprehensive approach to understanding AD and suggestions to be considered in the development of therapeutics for it are presented.
Collapse
|
19
|
The Interaction Between Contactin and Amyloid Precursor Protein and Its Role in Alzheimer’s Disease. Neuroscience 2020; 424:184-202. [DOI: 10.1016/j.neuroscience.2019.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 01/06/2023]
|
20
|
Sprooten J, Garg AD. Type I interferons and endoplasmic reticulum stress in health and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 350:63-118. [PMID: 32138904 PMCID: PMC7104985 DOI: 10.1016/bs.ircmb.2019.10.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFNs) comprise of pro-inflammatory cytokines created, as well as sensed, by all nucleated cells with the main objective of blocking pathogens-driven infections. Owing to this broad range of influence, type I IFNs also exhibit critical functions in many sterile inflammatory diseases and immunopathologies, especially those associated with endoplasmic reticulum (ER) stress-driven signaling pathways. Indeed, over the years accumulating evidence has indicated that the presence of ER stress can influence the production, or sensing of, type I IFNs induced by perturbations like pattern recognition receptor (PRR) agonists, infections (bacterial, viral or parasitic) or autoimmunity. In this article we discuss the link between type I IFNs and ER stress in various diseased contexts. We describe how ER stress regulates type I IFNs production or sensing, or how type I IFNs may induce ER stress, in various circumstances like microbial infections, autoimmunity, diabetes, cancer and other ER stress-related contexts.
Collapse
Affiliation(s)
- Jenny Sprooten
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium.
| |
Collapse
|
21
|
Sliker BH, Goetz BT, Peters HL, Poelaert BJ, Borgstahl GEO, Solheim JC. Beta 2-microglobulin regulates amyloid precursor-like protein 2 expression and the migration of pancreatic cancer cells. Cancer Biol Ther 2019; 20:931-940. [PMID: 30810435 DOI: 10.1080/15384047.2019.1580414] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Beta 2-microglobulin (β2m) is a component of the major histocompatibility complex (MHC) class I molecule, which presents tumor antigens to T lymphocytes to trigger cancer cell destruction. Notably, β2m has been reported as persistently expressed, rather than down regulated, in some tumor types. For renal cell and oral squamous cell carcinomas, β2m expression has been linked to increased migratory capabilities. The migratory ability of pancreatic cancer cells contributes to their metastatic tendencies and lethal nature. Therefore, in this study, we examined the impact of β2m on pancreatic cancer cell migration. We found that β2m protein is amply expressed in several human pancreatic cancer cell lines (S2-013, PANC-1, and MIA PaCa-2). Reducing β2m expression by short interfering RNA (siRNA) transfection significantly slowed the migration of the PANC-1 and S2-013 cancer cell lines, but increased the migration of the MIA PaCa-2 cell line. The amyloid precursor-like protein 2 (APLP2) has been documented as contributing to pancreatic cancer cell migration, invasiveness, and metastasis. We have previously shown that β2m/HLA class I/peptide complexes associate with APLP2 in S2-013 cells, and in this study we also detected their association in PANC-1 cells but not MIA PaCa-2 cells. In addition, siRNA down regulation of β2m expression diminished the expression of APLP2 in S2-013 and PANC-1 but heightened the level of APLP2 in MIA PaCa-2 cells, consistent with our migration data and co-immunoprecipitation data. Thus, our findings indicate that β2m regulates pancreatic cancer cell migration, and furthermore suggest that APLP2 is an intermediary in this process.
Collapse
Affiliation(s)
- Bailee H Sliker
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA
| | - Benjamin T Goetz
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA
| | - Haley L Peters
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA
| | - Brittany J Poelaert
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA
| | - Gloria E O Borgstahl
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA.,c Department of Biochemistry and Molecular Biology , University of Nebraska Medical Center , Omaha , NE , USA.,d Department of Pharmaceutical Sciences , University of Nebraska Medical Center , Omaha , NE , USA
| | - Joyce C Solheim
- a Eppley Institute for Research in Cancer and Allied Diseases , University of Nebraska Medical Center , Omaha , NE , USA.,b Fred and Pamela Buffett Cancer Center , University of Nebraska Medical Center , Omaha , NE , USA.,c Department of Biochemistry and Molecular Biology , University of Nebraska Medical Center , Omaha , NE , USA.,e Department of Pathology and Microbiology , University of Nebraska Medical Center , Omaha , NE , USA
| |
Collapse
|
22
|
|
23
|
O’Neill BT, Beck EM, Butler CR, Nolan CE, Gonzales C, Zhang L, Doran SD, Lapham K, Buzon LM, Dutra JK, Barreiro G, Hou X, Martinez-Alsina LA, Rogers BN, Villalobos A, Murray JC, Ogilvie K, LaChapelle EA, Chang C, Lanyon LF, Steppan CM, Robshaw A, Hales K, Boucher GG, Pandher K, Houle C, Ambroise CW, Karanian D, Riddell D, Bales KR, Brodney MA. Design and Synthesis of Clinical Candidate PF-06751979: A Potent, Brain Penetrant, β-Site Amyloid Precursor Protein Cleaving Enzyme 1 (BACE1) Inhibitor Lacking Hypopigmentation. J Med Chem 2018; 61:4476-4504. [DOI: 10.1021/acs.jmedchem.8b00246] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Brian T. O’Neill
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Elizabeth M. Beck
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Christopher R. Butler
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Charles E. Nolan
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Cathleen Gonzales
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Lei Zhang
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Shawn D. Doran
- Medicine Design, Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Kimberly Lapham
- Medicine Design, Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Leanne M. Buzon
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Jason K. Dutra
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Gabriela Barreiro
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Xinjun Hou
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | | | - Bruce N. Rogers
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Anabella Villalobos
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - John C. Murray
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Kevin Ogilvie
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Erik A. LaChapelle
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Cheng Chang
- Medicine Design, Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Lorraine F. Lanyon
- Discovery Sciences, Primary Pharmacology, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Claire M. Steppan
- Discovery Sciences, Primary Pharmacology, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Ashley Robshaw
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Katherine Hales
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Germaine G. Boucher
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Karamjeet Pandher
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Christopher Houle
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Claude W. Ambroise
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - David Karanian
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340, United States
| | - David Riddell
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Kelly R. Bales
- Internal Medicine, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| | - Michael A. Brodney
- Medicine Design, Medicinal Chemistry, Pfizer Inc., Cambridge, Massachusetts 02139, United States
| |
Collapse
|
24
|
Sharoar MG, Yan R. Effects of altered RTN3 expression on BACE1 activity and Alzheimer's neuritic plaques. Rev Neurosci 2018; 28:145-154. [PMID: 27883331 DOI: 10.1515/revneuro-2016-0054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022]
Abstract
Reticulon 3 (RTN3), which is a member of the reticulon family of proteins, has a biochemical function of shaping tubular endoplasmic reticulum. RTN3 has also been found to interact with β-site amyloid precursor protein cleaving enzyme 1 (BACE1), which initiates the generation of β-amyloid peptides (Aβ) from amyloid precursor protein. Aβ is the major proteinaceous component in neuritic plaques, which constitute one of the major pathological features in brains of Alzheimer's disease (AD) patients. Mice deficient in or overexpressing RTN3 have altered amyloid deposition through effects on BACE1 expression and activity. In this review, we will summarize the current findings concerning the role of RTN3 in AD pathogenesis and demonstrate that RTN3 protein levels act as age-dependent modulators of BACE1 activity and Aβ deposition during the pathogenic progression of AD.
Collapse
|
25
|
Hashimoto M, Ho G, Sugama S, Takamatsu Y, Shimizu Y, Takenouchi T, Waragai M, Masliah E. Evolvability of Amyloidogenic Proteins in Human Brain. J Alzheimers Dis 2018; 62:73-83. [PMID: 29439348 PMCID: PMC5817905 DOI: 10.3233/jad-170894] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2017] [Indexed: 12/29/2022]
Abstract
Currently, the physiological roles of amyloidogenic proteins (APs) in human brain, such as amyloid-β and α-synuclein, are elusive. Given that many APs arose by gene duplication and have been resistant against the pressures of natural selection, APs may be associated with some functions that are advantageous for survival of offspring. Nonetheless, evolvability is the sole physiological quality of APs that has been characterized in microorganisms such as yeast. Since yeast and human brain may share similar strategies in coping with diverse range of critical environmental stresses, the objective of this paper was to discuss the potential role of evolvability of APs in aging-associated neurodegenerative disorders, including Alzheimer's disease and Parkinson's disease. Given the heterogeneity of APs in terms of structure and cytotoxicity, it is argued that APs might be involved in preconditioning against diverse stresses in human brain. It is further speculated that these stress-related APs, most likely protofibrillar forms, might be transmitted to offspring via the germline, conferring preconditioning against forthcoming stresses. Thus, APs might represent a vehicle for the inheritance of the acquired characteristics against environmental stresses. Curiously, such a characteristic of APs is reminiscent of Charles Darwin's 'gemmules', imagined molecules of heritability described in his pangenesis theory. We propose that evolvability might be a physiological function of APs during the reproductive stage and neurodegenerative diseases could be a by-product effect manifested later in aging. Collectively, our evolvability hypothesis may play a complementary role in the pathophysiology of APs with the conventional amyloid cascade hypothesis.
Collapse
Affiliation(s)
- Makoto Hashimoto
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, USA
| | - Shuei Sugama
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Yoshiki Takamatsu
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Yuka Shimizu
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Masaaki Waragai
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Eliezer Masliah
- Division of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Islam MI, Sharoar MG, Ryu EK, Park IS. Limited activation of the intrinsic apoptotic pathway plays a main role in amyloid-β-induced apoptosis without eliciting the activation of the extrinsic apoptotic pathway. Int J Mol Med 2017; 40:1971-1982. [PMID: 29039468 DOI: 10.3892/ijmm.2017.3193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/03/2017] [Indexed: 11/06/2022] Open
Abstract
Amyloid-β (Aβ), a main pathogenic factor of Alzheimer's disease (AD), induces apoptosis accompanied by caspase activation. However, limited caspase activation and the suppression of the intrinsic apoptotic pathway (IAPW) are frequently observed upon Aβ treatment. In this study, we investigated whether these suppressive effects of Aβ can be overcome; we also examined the death-related pathways. Single treatments of cells with Aβ42 for up to 48 h barely induced caspase activation. In cells treated with Aβ42 twice for 2 h followed by 22 h (2+22 h) or for longer durations, the apoptotic protease activating factor-1 (Apaf-1) apoptosome was formed and caspases-3 and -9 were activated to a certain extent, suggesting the activation of the IAPW. However, the Aβ42-induced activation of the IAPW differed from that induced by treatment with other agents, such as staurosporine (STS) in that lower amounts of cytochrome c were released from the mitochondria, the majority of procaspase-9 in the Apaf-1 complex was not processed and caspase-3 was activated to a lesser extent in the peptide-treated cells. Thus, it seemed that the IAPW was not fully activated by Aβ42. The 30- and 41/43-kDa fragments derived from procaspase-8 were detected, which appear to be produced through the IAPW without death-inducing signaling-complex (DISC) formation, a key feature of the extrinsic apoptotic pathway (EAPW). Bid cleavage was observed only after caspase-3 activity reached its maximal levels, suggesting that the cleavage may contribute in a limited capacity to the amplification process of the IAPW in the Aβ-treated cells. Taken together, our data suggest that the IAPW, albeit functional only to a limited extent, plays a major role in Aβ42-induced apoptosis without the EAPW.
Collapse
Affiliation(s)
- Md Imamul Islam
- Department of Medical Sciences, Chosun University, Gwangju 501-759, Republic of Korea
| | - Md Golam Sharoar
- Department of Medical Sciences, Chosun University, Gwangju 501-759, Republic of Korea
| | - Eun-Kyoung Ryu
- Department of Nursing, Kongju National University, Kongju 314-701, Republic of Korea
| | - Il-Seon Park
- Department of Medical Sciences, Chosun University, Gwangju 501-759, Republic of Korea
| |
Collapse
|
27
|
Identification of additional loci associated with antibody response to Mycobacterium avium ssp. Paratuberculosis in cattle by GSEA-SNP analysis. Mamm Genome 2017; 28:520-527. [PMID: 28864882 DOI: 10.1007/s00335-017-9714-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 08/27/2017] [Indexed: 10/18/2022]
Abstract
Mycobacterium avium subsp. paratuberculosis: (MAP) causes a contagious chronic infection results in Johne's disease in a wide range of animal species, including cattle. Several genome-wide association studies (GWAS) have been carried out to identify loci putatively associated with MAP susceptibility by testing each marker separately and identifying SNPs that show a significant association with the phenotype, while SNP with modest effects are usually ignored. The objective of this study was to identify modest-effect genes associated with MAP susceptibility using a pathway-based approach. The Illumina BovineSNP50 BeadChip was used to genotype 966 Holstein cows, 483 positive and 483 negative for antibody response to MAP, data were then analyzed using novel SNP-based Gene Set Enrichment Analysis (GSEA-SNP) and validated with Adaptive Rank Truncated Product methodology. An allele-based test was carried out to estimate the statistical association for each marker with the phenotype, subsequently SNPs were mapped to the closest genes, considering for each gene the single variant with the highest value within a window of 50 kb, then pathway-statistics were tested using the GSEA-SNP method. The GO biological process "embryogenesis and morphogenesis" was most highly associated with antibody response to MAP. Within this pathway, five genes code for proteins which play a role in the immune defense relevant to response to bacterial infection. The immune response genes identified would not have been considered using a standard GWAS, thus demonstrating that the pathway approach can extend the interpretation of genome-wide association analyses and identify additional candidate genes for target traits.
Collapse
|
28
|
Han K, Müller UC, Hülsmann S. Amyloid-precursor Like Proteins APLP1 and APLP2 Are Dispensable for Normal Development of the Neonatal Respiratory Network. Front Mol Neurosci 2017; 10:189. [PMID: 28690498 PMCID: PMC5479907 DOI: 10.3389/fnmol.2017.00189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/29/2017] [Indexed: 11/13/2022] Open
Abstract
Recent studies using animal models indicated that the members of the amyloid precursor protein (APP) gene family are important for the formation, maintenance, and plasticity of synapses. Despite this, the specific role of the APP homologs APLP1 and APLP2 within the CNS and PNS is still poorly understood. In contrast to the subtle phenotypes of single mutants, double knockout mice (DKO) lacking APP/APLP2 or APLP1/APLP2 die within the first day after birth. Whereas APP/APLP2-DKO mice show severe deficits of neuromuscular morphology and transmission, the underlying cause of lethality of APLP1/APLP2-DKO mice remains unclear. Since expression of both proteins was confirmed by in situ hybridization, we aimed to test the role of APLP1/APLP2 in the formation and maintenance of synapses in the brainstem, and assessed a potential dysfunction of the most vital central neuronal network in APLP1/APLP2-DKO mice by analyzing the respiratory network of the medulla. We performed in vivo unrestrained whole body plethysmography in newborn APLP1/APLP2-DKO mice at postnatal day zero. Additionally, we directly tested the activity of the respiratory network in an acute slice preparation that includes the pre-Bötzinger complex. In both sets of experiments, no significant differences were detected regarding respiratory rate and cycle variability, strongly arguing against central respiratory problems as the primary cause of death of APLP1/APLP2-DKO mice. Thus, we conclude that APLP1 and APLP2 are dispensable for the development of the network and the generation of a normal breathing rhythm.
Collapse
Affiliation(s)
- Kang Han
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg UniversityHeidelberg, Germany
| | - Ulrike C Müller
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg UniversityHeidelberg, Germany
| | - Swen Hülsmann
- Klinik für Anästhesiologie, Universitätsmedizin GöttingenGöttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB)Göttingen, Germany
| |
Collapse
|
29
|
APLP1 Is a Synaptic Cell Adhesion Molecule, Supporting Maintenance of Dendritic Spines and Basal Synaptic Transmission. J Neurosci 2017; 37:5345-5365. [PMID: 28450540 DOI: 10.1523/jneurosci.1875-16.2017] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 02/22/2017] [Accepted: 03/24/2017] [Indexed: 12/19/2022] Open
Abstract
The amyloid precursor protein (APP), a key player in Alzheimer's disease, belongs to the family of synaptic adhesion molecules (SAMs) due to its impact on synapse formation and synaptic plasticity. These functions are mediated by both the secreted APP ectodomain that acts as a neurotrophic factor and full-length APP forming trans-cellular dimers. Two homologs of APP exist in mammals: the APP like proteins APLP1 and APLP2, exhibiting functions that partly overlap with those of APP. Here we tested whether APLP1 and APLP2 also show features of SAMs. We found that all three family members were upregulated during postnatal development coinciding with synaptogenesis. We observed presynaptic and postsynaptic localization of all APP family members and could show that heterologous expression of APLP1 or APLP2 in non-neuronal cells induces presynaptic differentiation in contacting axons of cocultured neurons, similar to APP and other SAMs. Moreover, APP/APLPs all bind to synaptic-signaling molecules, such as MINT/X11. Furthermore, we report that aged APLP1 knock-out mice show impaired basal transmission and a reduced mEPSC frequency, likely resulting from reduced spine density. This demonstrates an essential nonredundant function of APLP1 at the synapse. Compared to APP, APLP1 exhibits increased trans-cellular binding and elevated cell-surface levels due to reduced endocytosis. In conclusion, our results establish that APLPs show typical features of SAMs and indicate that increased surface expression, as observed for APLP1, is essential for proper synapse formation in vitro and synapse maintenance in vivoSIGNIFICANCE STATEMENT According to the amyloid-cascade hypothesis, Alzheimer's disease is caused by the accumulation of Aβ peptides derived from sequential cleavage of the amyloid precursor protein (APP) by β-site APP cleaving enzyme 1 (BACE1) and γ-secretase. Here we show that all mammalian APP family members (APP, APLP1, and APLP2) exhibit synaptogenic activity, involving trans-synaptic dimerization, similar to other synaptic cell adhesion molecules, such as Neuroligin/Neurexin. Importantly, our study revealed that the loss of APLP1, which is one of the major substrates of BACE1, causes reduced spine density in aged mice. Because some therapeutic interventions target APP processing (e.g., BACE inhibitors), those strategies may alter APP/APLP physiological function. This should be taken into account for the development of pharmaceutical treatments of Alzheimer's disease.
Collapse
|
30
|
Wild K, August A, Pietrzik CU, Kins S. Structure and Synaptic Function of Metal Binding to the Amyloid Precursor Protein and its Proteolytic Fragments. Front Mol Neurosci 2017; 10:21. [PMID: 28197076 PMCID: PMC5281630 DOI: 10.3389/fnmol.2017.00021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/16/2017] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is ultimately linked to the amyloid precursor protein (APP). However, current research reveals an important synaptic function of APP and APP-like proteins (APLP1 and 2). In this context various neurotrophic and neuroprotective functions have been reported for the APP proteolytic fragments sAPPα, sAPPβ and the monomeric amyloid-beta peptide (Aβ). APP is a metalloprotein and binds copper and zinc ions. Synaptic activity correlates with a release of these ions into the synaptic cleft and dysregulation of their homeostasis is linked to different neurodegenerative diseases. Metal binding to APP or its fragments affects its structure and its proteolytic cleavage and therefore its physiological function at the synapse. Here, we summarize the current data supporting this hypothesis and provide a model of how these different mechanisms might be intertwined with each other.
Collapse
Affiliation(s)
- Klemens Wild
- Heidelberg University Biochemistry Center (BZH), University of Heidelberg Heidelberg, Germany
| | - Alexander August
- Division of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Claus U Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz Mainz, Germany
| | - Stefan Kins
- Division of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| |
Collapse
|
31
|
Ludewig S, Korte M. Novel Insights into the Physiological Function of the APP (Gene) Family and Its Proteolytic Fragments in Synaptic Plasticity. Front Mol Neurosci 2017; 9:161. [PMID: 28163673 PMCID: PMC5247455 DOI: 10.3389/fnmol.2016.00161] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 12/14/2016] [Indexed: 12/31/2022] Open
Abstract
The amyloid precursor protein (APP) is well known to be involved in the pathophysiology of Alzheimer's disease (AD) via its cleavage product amyloid ß (Aß). However, the physiological role of APP, its various proteolytic products and the amyloid precursor-like proteins 1 and 2 (APLP1/2) are still not fully clarified. Interestingly, it has been shown that learning and memory processes represented by functional and structural changes at synapses are altered in different APP and APLP1/2 mouse mutants. In addition, APP and its fragments are implicated in regulating synaptic strength further reinforcing their modulatory role at the synapse. While APLP2 and APP are functionally redundant, the exclusively CNS expressed APLP1, might have individual roles within the synaptic network. The proteolytic product of non-amyloidogenic APP processing, APPsα, emerged as a neurotrophic peptide that facilitates long-term potentiation (LTP) and restores impairments occurring with age. Interestingly, the newly discovered η-secretase cleavage product, An-α acts in the opposite direction, namely decreasing LTP. In this review we summarize recent findings with emphasis on the physiological role of the APP gene family and its proteolytic products on synaptic function and plasticity, especially during processes of hippocampal LTP. Therefore, we focus on literature that provide electrophysiological data by using different mutant mouse strains either lacking full-length or parts of the APP proteins or that utilized secretase inhibitors as well as secreted APP fragments.
Collapse
Affiliation(s)
- Susann Ludewig
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU BraunschweigBraunschweig, Germany; Helmholtz Centre for Infection Research, AG NINDBraunschweig, Germany
| |
Collapse
|
32
|
The multifaceted role of metalloproteinases in physiological and pathological conditions in embryonic and adult brains. Prog Neurobiol 2016; 155:36-56. [PMID: 27530222 DOI: 10.1016/j.pneurobio.2016.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 07/10/2016] [Accepted: 08/08/2016] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinases (MMPs) are a large family of ubiquitous extracellular endopeptidases, which play important roles in a variety of physiological and pathological conditions, from the embryonic stages throughout adult life. Their extraordinary physiological "success" is due to concomitant broad substrate specificities and strict regulation of their expression, activation and inhibition levels. In recent years, MMPs have gained increasing attention as significant effectors in various aspects of central nervous system (CNS) physiology. Most importantly, they have been recognized as main players in a variety of brain disorders having different etiologies and evolution. A common aspect of these pathologies is the development of acute or chronic neuroinflammation. MMPs play an integral part in determining the result of neuroinflammation, in some cases turning its beneficial outcome into a harmful one. This review summarizes the most relevant studies concerning the physiology of MMPs, highlighting their involvement in both the developing and mature CNS, in long-lasting and acute brain diseases and, finally, in nervous system repair. Recently, a concerted effort has been made in identifying therapeutic strategies for major brain diseases by targeting MMP activities. However, from this revision of the literature appears clear that MMPs have multifaceted functional characteristics, which modulate physiological processes in multiple ways and with multiple consequences. Therefore, when choosing MMPs as possible targets, great care must be taken to evaluate the delicate balance between their activation and inhibition and to determine at which stage of the disease and at what level they become active in order maximize chances of success.
Collapse
|
33
|
Guntupalli S, Widagdo J, Anggono V. Amyloid-β-Induced Dysregulation of AMPA Receptor Trafficking. Neural Plast 2016; 2016:3204519. [PMID: 27073700 PMCID: PMC4814684 DOI: 10.1155/2016/3204519] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/28/2016] [Indexed: 01/22/2023] Open
Abstract
Evidence from neuropathological, genetic, animal model, and biochemical studies has indicated that the accumulation of amyloid-beta (Aβ) is associated with, and probably induces, profound neuronal changes in brain regions critical for memory and cognition in the development of Alzheimer's disease (AD). There is considerable evidence that synapses are particularly vulnerable to AD, establishing synaptic dysfunction as one of the earliest events in pathogenesis, prior to neuronal loss. It is clear that excessive Aβ levels can disrupt excitatory synaptic transmission and plasticity, mainly due to dysregulation of the AMPA and NMDA glutamate receptors in the brain. Importantly, AMPA receptors are the principal glutamate receptors that mediate fast excitatory neurotransmission. This is essential for synaptic plasticity, a cellular correlate of learning and memory, which are the cognitive functions that are most disrupted in AD. Here we review recent advances in the field and provide insights into the molecular mechanisms that underlie Aβ-induced dysfunction of AMPA receptor trafficking. This review focuses primarily on NMDA receptor- and metabotropic glutamate receptor-mediated signaling. In particular, we highlight several mechanisms that underlie synaptic long-term depression as common signaling pathways that are hijacked by the neurotoxic effects of Aβ.
Collapse
Affiliation(s)
- Sumasri Guntupalli
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
34
|
Abstract
Here we review the similarities between a rare inherited disorder, familial British dementia (FBD), and the most common of all late-life neurological conditions, Alzheimer's diseases (AD). We describe the symptoms, pathology and genetics of FBD, the biology of the BRI2 protein and mouse models of FBD and familial Danish dementia. In particular, we focus on the evolving recognition of the importance of protein oligomers and aberrant processing of the amyloid β-protein precursor (APP) - themes that are common to both FBD and AD. The initial discovery that FBD is phenotypically similar to AD, but associated with the deposition of an amyloid peptide (ABri) distinct from the amyloid β-protein (Aβ) led many to assume that amyloid production alone is sufficient to initiate disease and that ABri is the molecular equivalent of Aβ. Parallel with work on Aβ, studies of ABri producing animal models and in vitro ABri toxicity experiments caused a revision of the amyloid hypothesis and a focus on soluble oligomers of Aβ and ABri. Contemporaneous other studies suggested that loss of the ABri precursor protein (BRI2) may underlie the cognitive deficits in FBD. In this regard it is important to note that BRI2 has been shown to interact with and regulate the processing of APP, and that mutant BRI2 leads to altered cleavage of APP. A synthesis of these results suggests that a “two-hit mechanism” better explains FBD than earlier toxic gain of function and toxic loss of function models. The lessons learned from the study of FBD imply that the molecular pathology of AD is also likely to involve both aberrant aggregation (in AD, Aβ) and altered APP processing. With regard to FBD, we propose that the C-terminal 11 amino acid of FBD-BRI2 interfere with both the normal function of BRI2 and promotes the production of cystine cross-linked toxic ABri oligomers. In this scenario, loss of BRI2 function leads to altered APP processing in as yet underappreciated ways. Given the similarities between FBD and AD it seems likely that study of the structure of ABri oligomers and FBD-induced changes in APP metabolites will further our understanding of AD.
Collapse
Affiliation(s)
- Adam Cantlon
- Laboratory for Neurodegenerative Research, School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Republic of Ireland ; Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - Carlo Sala Frigerio
- Laboratory for Neurodegenerative Research, School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Republic of Ireland
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| |
Collapse
|
35
|
Hua R, Wei M, Zhang C. The complex genetics in autism spectrum disorders. SCIENCE CHINA-LIFE SCIENCES 2015; 58:933-45. [PMID: 26335739 DOI: 10.1007/s11427-015-4893-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Autism spectrum disorders (ASD) are a pervasive neurodevelopmental disease characterized by deficits in social interaction and nonverbal communication, as well as restricted interests and stereotypical behavior. Genetic changes/heritability is one of the major contributing factors, and hundreds to thousands of causative and susceptible genes, copy number variants (CNVs), linkage regions, and microRNAs have been associated with ASD which clearly indicates that ASD is a complex genetic disorder. Here, we will briefly summarize some of the high-confidence genetic changes in ASD and their possible roles in their pathogenesis.
Collapse
Affiliation(s)
- Rui Hua
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - MengPing Wei
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Chen Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
36
|
Singh D, Srivastava SK, Chaudhuri TK, Upadhyay G. Multifaceted role of matrix metalloproteinases (MMPs). Front Mol Biosci 2015; 2:19. [PMID: 25988186 PMCID: PMC4429632 DOI: 10.3389/fmolb.2015.00019] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/28/2015] [Indexed: 12/31/2022] Open
Abstract
Matrix metalloproteinases (MMPs), a large family of calcium-dependent zinc-containing endopeptidases, are involved in the tissue remodeling and degradation of the extracellular matrix. MMPs are widely distributed in the brain and regulate various processes including microglial activation, inflammation, dopaminergic apoptosis, blood-brain barrier disruption, and modulation of α-synuclein pathology. High expression of MMPs is well documented in various neurological disorders including Parkinson's disease (PD), Alzheimer's disease (AD), Japanese encephalitis (JE), and Glaucoma. Although potentially critical, the role of MMPs in neuronal disorders is under-investigated. The present review summarizes the role of MMPs in neurodegeneration with a particular emphasis on PD, AD, JE, and Glaucoma.
Collapse
Affiliation(s)
- Divya Singh
- Department of Biology, City College of New York New York, NY, USA
| | - Sanjeev K Srivastava
- Cellular Immunology Laboratory, Department of Zoology, University of North Bengal Siliguri, India
| | - Tapas K Chaudhuri
- Cellular Immunology Laboratory, Department of Zoology, University of North Bengal Siliguri, India
| | | |
Collapse
|
37
|
Contactin-4 mediates axon-target specificity and functional development of the accessory optic system. Neuron 2015; 86:985-999. [PMID: 25959733 DOI: 10.1016/j.neuron.2015.04.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 02/19/2015] [Accepted: 03/31/2015] [Indexed: 12/31/2022]
Abstract
The mammalian eye-to-brain pathway includes more than 20 parallel circuits, each consisting of precise long-range connections between specific sets of retinal ganglion cells (RGCs) and target structures in the brain. The mechanisms that drive assembly of these parallel connections and the functional implications of their specificity remain unresolved. Here we show that in the absence of contactin 4 (CNTN4) or one of its binding partners, amyloid precursor protein (APP), a subset of direction-selective RGCs fail to target the nucleus of the optic tract (NOT)--the accessory optic system (AOS) target controlling horizontal image stabilization. Conversely, ectopic expression of CNTN4 biases RGCs to arborize in the NOT, and that process also requires APP. Our data reveal critical and novel roles for CNTN4/APP in promoting target-specific axon arborization, and they highlight the importance of this process for functional development of a behaviorally relevant parallel visual pathway.
Collapse
|
38
|
Kulikova AA, Makarov AA, Kozin SA. Roles of zinc ions and structural polymorphism of β-amyloid in the development of Alzheimer’s disease. Mol Biol 2015. [DOI: 10.1134/s0026893315020065] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
39
|
Vnencak M, Paul MH, Hick M, Schwarzacher SW, Del Turco D, Müller UC, Deller T, Jedlicka P. Deletion of the amyloid precursor-like protein 1 (APLP1) enhances excitatory synaptic transmission, reduces network inhibition but does not impair synaptic plasticity in the mouse dentate gyrus. J Comp Neurol 2015; 523:1717-29. [PMID: 25728909 DOI: 10.1002/cne.23766] [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: 11/10/2014] [Revised: 02/21/2015] [Accepted: 02/23/2015] [Indexed: 01/02/2023]
Abstract
Amyloid precursor-like protein 1 (APLP1) is a transmembrane synaptic protein belonging to the amyloid precursor protein (APP) gene family. Although the role of this gene family-in particular of APP-has been intensely studied in the context of Alzheimer's disease, the physiological roles of its family members remain poorly understood. In particular, the function of APLP1, which is predominantly expressed in the nervous system, has remained enigmatic. Since APP has been implicated in synaptic plasticity, we wondered whether APLP1 could play a similar role. First, using in situ hybridization and laser microdissection combined with reverse transcription-quantitative polymerase chain reaction (PCR) we observed that Aplp1 mRNA is highly expressed in dentate granule cells. Having this examined, we studied synaptic plasticity at the perforant path-granule cell synapses in the dentate gyrus of APLP1-deficient mice in vivo. Analysis of field excitatory postsynaptic potentials evoked by stimulation of perforant path fibers revealed increased excitatory transmission in APLP1-deficient mice. Moreover, we observed decreased paired-pulse inhibition of population spikes indicating a decrease in network inhibition upon deletion of APLP1. In contrast, short-term presynaptic plasticity (STP) as well as long-term synaptic plasticity (LTP) was unchanged in the absence of APLP1. Based on these results we conclude that APLP1 deficiency on its own does not lead to defects in synaptic plasticity, but affects synaptic transmission and network inhibition in the dentate gyrus.
Collapse
Affiliation(s)
- Matej Vnencak
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt am Main, Germany
| | - Mandy H Paul
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt am Main, Germany
| | - Meike Hick
- Department of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Stephan W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt am Main, Germany
| | - Domenico Del Turco
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt am Main, Germany
| | - Ulrike C Müller
- Department of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt am Main, Germany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe-University, Frankfurt am Main, Germany
| |
Collapse
|
40
|
Shamsuzzama, Kumar L, Haque R, Nazir A. Role of MicroRNA Let-7 in Modulating Multifactorial Aspect of Neurodegenerative Diseases: an Overview. Mol Neurobiol 2015; 53:2787-2793. [PMID: 25823513 DOI: 10.1007/s12035-015-9145-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/18/2015] [Indexed: 10/23/2022]
Abstract
The multifactorial aspect of neurodegenerative diseases has posed challenges in terms of understanding various mechanistic cues behind these ailments. The fact that single microRNA (miRNA) molecules can regulate multiple genes and associated pathways makes these molecules interesting for studies within the area of age-associated neurodegenerative diseases. miRNAs are endogenous, evolutionarily conserved, 20-23 nucleotide non-coding RNAs, which were first discovered in Caenorhabditis elegans. They play a key role in gene regulation and are known to be deregulated in many disease conditions. Steady regulations of miRNAs are required for normal biological processes. One of the crucial miRNA molecules let-7 is highly conserved and is known to be required for development and viability. It acts as a regulator for oncogenes and insulin-PI3K-mTOR pathway genes. Upregulation of let-7 impairs glucose homeostasis and results in degeneration of neurons, while its downregulation leads to cancer. Maturation of let-7 in cancer subjects is inhibited by lin-28, an RNA-binding protein inhibitor. This highlights the importance of let-7 miRNAs in various diseases and developmental processes. This article provides an overview on the functions of let-7 and its probable association with various neurodegenerative diseases.
Collapse
Affiliation(s)
- Shamsuzzama
- Laboratory of Functional Genomics and Molecular Toxicology, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, UP, 226 031, India
| | - Lalit Kumar
- Laboratory of Functional Genomics and Molecular Toxicology, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, UP, 226 031, India
| | - Rizwanul Haque
- Laboratory of Functional Genomics and Molecular Toxicology, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, UP, 226 031, India
| | - Aamir Nazir
- Laboratory of Functional Genomics and Molecular Toxicology, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, UP, 226 031, India.
| |
Collapse
|
41
|
Brodney MA, Beck EM, Butler CR, Barreiro G, Johnson EF, Riddell D, Parris K, Nolan CE, Fan Y, Atchison K, Gonzales C, Robshaw AE, Doran SD, Bundesmann MW, Buzon L, Dutra J, Henegar K, LaChapelle E, Hou X, Rogers BN, Pandit J, Lira R, Martinez-Alsina L, Mikochik P, Murray JC, Ogilvie K, Price L, Sakya SM, Yu A, Zhang Y, O'Neill BT. Utilizing structures of CYP2D6 and BACE1 complexes to reduce risk of drug-drug interactions with a novel series of centrally efficacious BACE1 inhibitors. J Med Chem 2015; 58:3223-52. [PMID: 25781223 PMCID: PMC4415909 DOI: 10.1021/acs.jmedchem.5b00191] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
In recent years, the first generation
of β-secretase (BACE1)
inhibitors advanced into clinical development for the treatment of
Alzheimer’s disease (AD). However, the alignment of drug-like
properties and selectivity remains a major challenge. Herein, we describe
the discovery of a novel class of potent, low clearance, CNS penetrant
BACE1 inhibitors represented by thioamidine 5. Further
profiling suggested that a high fraction of the metabolism (>95%)
was due to CYP2D6, increasing the potential risk for victim-based
drug–drug interactions (DDI) and variable exposure in the clinic
due to the polymorphic nature of this enzyme. To guide future design,
we solved crystal structures of CYP2D6 complexes with substrate 5 and its corresponding metabolic product pyrazole 6, which provided insight into the binding mode and movements between
substrate/inhibitor complexes. Guided by the BACE1 and CYP2D6 crystal
structures, we designed and synthesized analogues with reduced risk
for DDI, central efficacy, and improved hERG therapeutic margins.
Collapse
Affiliation(s)
| | | | | | | | - Eric F Johnson
- #The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92024, United States
| | | | | | | | - Ying Fan
- #The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92024, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Aijia Yu
- ∇WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Yong Zhang
- ∇WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | | |
Collapse
|
42
|
Unno K, Konishi T, Nakagawa A, Narita Y, Takabayashi F, Okamura H, Hara A, Yamamoto H, Iguchi K, Hoshino M, Yasui K, Katayanagi Y, Fukutomi R, Imai S. Cognitive dysfunction and amyloid β accumulation are ameliorated by the ingestion of green soybean extract in aged mice. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
43
|
Pain C, Dumont J, Dumoulin M. Camelid single-domain antibody fragments: Uses and prospects to investigate protein misfolding and aggregation, and to treat diseases associated with these phenomena. Biochimie 2015; 111:82-106. [DOI: 10.1016/j.biochi.2015.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/23/2015] [Indexed: 12/19/2022]
|
44
|
Cousins SL, Dai W, Stephenson FA. APLP1 and APLP2, members of the APP family of proteins, behave similarly to APP in that they associate with NMDA receptors and enhance NMDA receptor surface expression. J Neurochem 2015; 133:879-85. [PMID: 25683482 DOI: 10.1111/jnc.13063] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/02/2015] [Accepted: 02/06/2015] [Indexed: 11/30/2022]
Abstract
The function of amyloid precursor protein (APP) is unknown, although the discovery that it contributes to the regulation of surface expression of N-methyl-D-aspartate (NMDA) receptors has afforded new insights into its functional significance. Since APP is a member of a gene family that contains two other members, amyloid precursor-like proteins 1 and 2 (APLP1 and APLP2), it is important to determine if the related APP proteins possess the same properties as APP with respect to their interactions with NMDA receptors. Following expression in mammalian cells, both APLP1 and APLP2 behaved similarly to APP in that they both co-immunoprecipitated with the two major NMDA receptor subtypes, GluN1/GluN2A and GluN1/GluN2B, via interaction with the obligatory GluN1 subunit. Immunoprecipitations from detergent extracts of adult mammalian brain showed co-immunoprecipitation of APLP1 and APLP2 with GluN2A- and GluN2B-containing NMDA receptors. Furthermore, similarly to APP, APLP1 and APLP2 both enhanced GluN1/GluN2A and GluN1/GluN2B cell surface expression. Thus, all the three members of the APP gene family behave similarly in that they each contribute to the regulation of cell surface NMDA receptor homoeostasis. Amyloid precursor protein (APP) has been shown to associate with N-methyl-d-aspartate (NMDA) receptors and to enhance their cell surface expression. Here, we show that the other members of the APP family, APLP1 and APLP2, behave similarly to APP in that they both associate with assembled NMDA receptors in the endoplasmic reticulum via their interaction with the NMDA receptor subunit, GluN1 and, they enhance receptor cell surface expression. Alternative scenarios are depicted since it is to be determined if respective associations are direct.
Collapse
Affiliation(s)
| | - Wei Dai
- University College London School of Pharmacy, London, UK
| | | |
Collapse
|
45
|
Cheng F, Cappai R, Lidfeldt J, Belting M, Fransson LÅ, Mani K. Amyloid precursor protein (APP)/APP-like protein 2 (APLP2) expression is required to initiate endosome-nucleus-autophagosome trafficking of glypican-1-derived heparan sulfate. J Biol Chem 2015; 289:20871-8. [PMID: 24898256 DOI: 10.1074/jbc.m114.552810] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Anhydromannose (anMan)-containing heparan sulfate (HS) derived from the proteoglycan glypican-1 is generated in endosomes by an endogenously or ascorbate-induced S-nitrosothiolcatalyzed reaction. Processing of the amyloid precursor protein (APP) and APP-like protein 2 (APLP2) by β- and γ-secretases into amyloid β(A) and Aβ-like peptides also takes place in these compartments. Moreover, anMan-containing HS suppresses the formation of toxic Aβ assemblies in vitro. We showed by using deconvolution immunofluorescence microscopy with an anMan-specific monoclonal antibody as well as (35)S labeling experiments that expression of APP/APLP2 is required for ascorbate-induced transport of HS from endosomes to the nucleus. Nuclear translocation was observed in wild-type mouse embryonic fibroblasts (WT MEFs), Tg2576 MEFs, and N2a neuroblastoma cells but not in APP(-/-) and APLP2(-/-) MEFs. Transfection of APP(-/-) cells with a vector encoding APP restored nuclear import of anMan-containing HS. In WT MEFs and N2a neuroblastoma cells exposed to β- or γ-secretase inhibitors, nuclear translocation was greatly impeded, suggesting involvement of APP/APLP2 degradation products. In Tg2576 MEFs, the β-inhibitor blocked transport, but the γ-inhibitor did not. During chase in ascorbate- free medium, anMan-containing HS disappeared from the nuclei of WT MEFs. Confocal immunofluorescence microscopy showed that they appeared in acidic, LC3-positive vesicles in keeping with an autophagosomal location. There was increased accumulation of anMan-containing HS in nuclei and cytosolic vesicles upon treatment with chloroquine, indicating that HS was degraded in lysosomes. Manipulations of APP expression and processing may have deleterious effects upon HS function in the nucleus.
Collapse
|
46
|
Hick M, Herrmann U, Weyer SW, Mallm JP, Tschäpe JA, Borgers M, Mercken M, Roth FC, Draguhn A, Slomianka L, Wolfer DP, Korte M, Müller UC. Acute function of secreted amyloid precursor protein fragment APPsα in synaptic plasticity. Acta Neuropathol 2015; 129:21-37. [PMID: 25432317 DOI: 10.1007/s00401-014-1368-x] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 11/24/2022]
Abstract
The key role of APP in the pathogenesis of Alzheimer disease is well established. However, postnatal lethality of double knockout mice has so far precluded the analysis of the physiological functions of APP and the APLPs in the brain. Previously, APP family proteins have been implicated in synaptic adhesion, and analysis of the neuromuscular junction of constitutive APP/APLP2 mutant mice showed deficits in synaptic morphology and neuromuscular transmission. Here, we generated animals with a conditional APP/APLP2 double knockout (cDKO) in excitatory forebrain neurons using NexCre mice. Electrophysiological recordings of adult NexCre cDKOs indicated a strong synaptic phenotype with pronounced deficits in the induction and maintenance of hippocampal LTP and impairments in paired pulse facilitation, indicating a possible presynaptic deficit. These deficits were also reflected in impairments in nesting behavior and hippocampus-dependent learning and memory tasks, including deficits in Morris water maze and radial maze performance. Moreover, while no gross alterations of brain morphology were detectable in NexCre cDKO mice, quantitative analysis of adult hippocampal CA1 neurons revealed prominent reductions in total neurite length, dendritic branching, reduced spine density and reduced spine head volume. Strikingly, the impairment of LTP could be selectively rescued by acute application of exogenous recombinant APPsα, but not APPsβ, indicating a crucial role for APPsα to support synaptic plasticity of mature hippocampal synapses on a rapid time scale. Collectively, our analysis reveals an essential role of APP family proteins in excitatory principal neurons for mediating normal dendritic architecture, spine density and morphology, synaptic plasticity and cognition.
Collapse
Affiliation(s)
- Meike Hick
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Stahl R, Schilling S, Soba P, Rupp C, Hartmann T, Wagner K, Merdes G, Eggert S, Kins S. Shedding of APP limits its synaptogenic activity and cell adhesion properties. Front Cell Neurosci 2014; 8:410. [PMID: 25520622 PMCID: PMC4253958 DOI: 10.3389/fncel.2014.00410] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 11/11/2014] [Indexed: 01/05/2023] Open
Abstract
The amyloid precursor protein (APP) plays a central role in Alzheimer's disease (AD) and has essential synapse promoting functions. Synaptogenic activity as well as cell adhesion properties of APP presumably depend on trans-cellular dimerization via its extracellular domain. Since neuronal APP is extensively processed by secretases, it raises the question if APP shedding affects its cell adhesion and synaptogenic properties. We show that inhibition of APP shedding using cleavage deficient forms of APP or a dominant negative α-secretase strongly enhanced its cell adhesion and synaptogenic activity suggesting that synapse promoting function of APP is tightly regulated by α-secretase mediated processing, similar to other trans-cellular synaptic adhesion molecules.
Collapse
Affiliation(s)
- Ronny Stahl
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Department of Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University Munich Munich, Germany
| | - Sandra Schilling
- Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Peter Soba
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Center for Molecular Neurobiology (ZMNH), University of Hamburg Hamburg, Germany
| | - Carsten Rupp
- Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Tobias Hartmann
- Deutsches Institut für DemenzPrävention, Experimental Neurology, Saarland University Homburg/Saar, Germany
| | - Katja Wagner
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Gunter Merdes
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Department of Biosystems Science and Engineering, ETH Zürich Basel, Switzerland
| | - Simone Eggert
- Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany
| | - Stefan Kins
- Center of Molecular Biology ZMBH, University of Heidelberg Heidelberg, Germany ; Department of Human Biology and Human Genetics, Technical University of Kaiserslautern Kaiserslautern, Germany ; Deutsches Institut für DemenzPrävention, Experimental Neurology, Saarland University Homburg/Saar, Germany
| |
Collapse
|
48
|
Mukherjee A, Swarnakar S. Implication of matrix metalloproteinases in regulating neuronal disorder. Mol Biol Rep 2014; 42:1-11. [DOI: 10.1007/s11033-014-3752-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
49
|
Marr RA, Hafez DM. Amyloid-beta and Alzheimer's disease: the role of neprilysin-2 in amyloid-beta clearance. Front Aging Neurosci 2014; 6:187. [PMID: 25165447 PMCID: PMC4131500 DOI: 10.3389/fnagi.2014.00187] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 07/09/2014] [Indexed: 12/14/2022] Open
Abstract
Accumulation of the amyloid-beta (Aβ) peptide is a central factor in Alzheimer's disease (AD) pathogenesis as supported by continuing evidence. This review concisely summarizes this evidence supporting a critical role for Aβ in AD before discussing the clearance of this peptide. Mechanisms of clearance of Aβ are critical for preventing pathological elevations in Aβ concentration. Direct degradation of Aβ by endopeptidases has emerged as one important pathway for clearance. Of particular interest are endopeptidases that are sensitive to the neprilysin (NEP) inhibitors thiorphan and phosphoramidon (i.e., are "NEP-like") as these inhibitors induce a dramatic increase in Aβ levels in rodents. This review will focus on neprilysin-2 (NEP2), a NEP-like endopeptidase which cooperates with NEP to control Aβ levels in the brain. The evidence for the involvement of NEP2 in AD is discussed as well as the therapeutic relevance with regards to gene therapy and the development of molecular markers for the disease.
Collapse
Affiliation(s)
- Robert A Marr
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science North Chicago, IL, USA
| | - Daniel M Hafez
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science North Chicago, IL, USA
| |
Collapse
|
50
|
Ciccotosto GD, James SA, Altissimo M, Paterson D, Vogt S, Lai B, de Jonge MD, Howard DL, Bush AI, Cappai R. Quantitation and localization of intracellular redox active metals by X-ray fluorescence microscopy in cortical neurons derived from APP and APLP2 knockout tissue. Metallomics 2014; 6:1894-904. [PMID: 25098278 DOI: 10.1039/c4mt00176a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The amyloid precursor protein (APP) gene family includes APP and the amyloid precursor-like proteins, APLP1 and APLP2. These proteins contain metal binding sites for copper, zinc and iron and are known to have physiological roles in modulating the metal homeostasis in brain cells. Here we report the application of X-ray fluorescence microscopy (XFM) to investigate the subcellular distribution patterns of the metal ions Cu, Zn, Fe, and Ca in individual neurons derived from APP and APLP2 knockout mice brains to further define their role in metal homeostasis. These studies add to the growing body of data that the APP family of proteins are metalloproteins that have shared as well as distinct effects on metals. As we continue to delineate the cellular effects of the APP family of proteins it is important to consider how metals are involved in their actions.
Collapse
Affiliation(s)
- Giuseppe D Ciccotosto
- Department of Pathology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|