1
|
Jia M, Wang C, Mei J, Ahmad S, Nouman MF, Ai H. Identification and Characterization of the Structure and Size of Aβ42 Oligomers Targeting the Receptor FcγRIIb. ACS Chem Neurosci 2025; 16:1335-1345. [PMID: 40094208 DOI: 10.1021/acschemneuro.4c00862] [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] [Indexed: 03/19/2025] Open
Abstract
Kam and colleagues discovered that FcγRIIb can specifically bind to Aβ42 oligomers (AβOs). The N-terminal residues F4 and D7 of Aβ42, as well as the W115 residue in domain D2 of FcγRIIb, are involved in this binding. However, the specificity of the FcγRIIb receptor's binding sites for AβOs and their dependence on different AβO species, including dimers (D/DT), trimers (T/TT), tetramers (Te/TeT), and pentamers (P/PT) during both the primary (P1) and secondary nucleation phases (P2), remains unknown. To address this, we employed molecular dynamics (MD) simulations to investigate the interactions between the extracellular domains D1 and D2 (FDD) of FcγRIIb and AβOs of varying sizes in the two different phases. We discovered that three specific fragments (f1, f2, and f3) of domain D2 in FDD are the primary binding sites for AβO species. Furthermore, among AβOs of the same molecular weight, those from the P2 phase exhibit a stronger binding affinity for FDD than those from the P1 phase. The distinction is ascribed to the stronger dependence on the hydrophobic residues in the β1 and β2 regions for the binding of AβOs in P2 (including TT, TeT, and PT) than that (including D, Te, and P) in the P1 phase. In the P1 phase, these AβOs prefer to achieve binding to FDD through their N-terminal residues; however, by this, we identified that the species observed in Kam's experiment to bind FcγRIIb should probably be the tetrameric AβO (Te) in the P1 phase. Moreover, within both the P1 and P2 phases, we predicted that the trimeric AβO species in either the P1 or P2 phase is the strongest binding ligand for the FcγRIIb receptor. This study provides a comprehensive molecular perspective on the interaction between FcγRIIb and AβO in P2, which is of significant importance for the development of therapeutic strategies targeting Alzheimer's disease (AD) and autoimmune diseases.
Collapse
Affiliation(s)
- Mengke Jia
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
- Zibo City Engineering Research Center for New Pollution Monitoring and Governance, Shandong Vocational College of Light Industry, Zibo 255300, Shandong, P. R. China
| | - Chuanbo Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Jinfei Mei
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Sajjad Ahmad
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Muhammad Fahad Nouman
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Hongqi Ai
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| |
Collapse
|
2
|
Burns AP, Fortel I, Zhan L, Lazarov O, Mackin RS, Demos AP, Bendlin B, Leow A. Longitudinal excitation-inhibition balance altered by sex and APOE-ε4. Commun Biol 2025; 8:488. [PMID: 40133608 PMCID: PMC11937384 DOI: 10.1038/s42003-025-07876-5] [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: 09/04/2024] [Accepted: 03/03/2025] [Indexed: 03/27/2025] Open
Abstract
Neuronal hyperexcitation affects memory and neural processing across the Alzheimer's disease (AD) cognitive continuum. Levetiracetam, an antiepileptic, shows promise in improving cognitive impairment by restoring the neural excitation/inhibition balance in AD patients. We previously identified a hyper-excitable phenotype in cognitively unimpaired female APOE-ε4 carriers relative to male counterparts cross-sectionally. This sex difference lacks longitudinal validation; however, clarifying the vulnerability of female ε4-carriers could better inform antiepileptic treatment efficacy. Here, we investigated this sex-by-ε4 interaction using a longitudinal design. We used resting-state fMRI and diffusion tensor imaging collected longitudinally from 106 participants who were cognitively unimpaired for at least one scan event but may have been assessed to have clinical dementia ratings corresponding to early mild cognitive impairment over time. By including scan events where participants transitioned to mild cognitive impairment, we modeled the trajectory of the whole-brain excitation-inhibition ratio throughout the preclinical cognitively healthy continuum and extended to early impairment. A linear mixed model revealed a significant three-way interaction among sex, ε4-status, and time, with female ε4-carriers showing a significant hyper-excitable trajectory. These findings suggest a possible pathway for preventative therapy targeting preclinical hyperexcitation in female ε4-carriers.
Collapse
Affiliation(s)
- Andrew P Burns
- Department of Biomedical Engineering University of Illinois Chicago (UIC), 851 S Morgan St, Chicago, IL, 60607, USA.
| | - Igor Fortel
- Department of Biomedical Engineering University of Illinois Chicago (UIC), 851 S Morgan St, Chicago, IL, 60607, USA
| | - Liang Zhan
- Department of Electrical and Computer Engineering, University of Pittsburgh, 4200 Fifth Avenue, Pittsburgh, PA, 15260, USA
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois Chicago, 808 S. Wood St, Chicago, IL, 60612, USA
| | - R Scott Mackin
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, 675 18th St, San Francisco, CA, 94107, USA
- Department of Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA, USA
| | - Alexander P Demos
- Department of Psychology, University of Illinois Chicago (UIC), 1007 W Harrison St, Chicago, IL, 60607, USA
| | - Barbara Bendlin
- Department of Medicine, University of Wisconsin-Madison, 5158 Medical Foundation Centennial Building, 1685 Highland Ave, Madison, WI, 53792, USA
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, 600 Highland Ave J5/1 Mezzanine, Madison, WI, 53792, USA
| | - Alex Leow
- Department of Biomedical Engineering University of Illinois Chicago (UIC), 851 S Morgan St, Chicago, IL, 60607, USA.
| |
Collapse
|
3
|
Arruda BP, Martins PP, Kihara AH, Takada SH. Perinatal asphyxia and Alzheimer's disease: is there a correlation? Front Pediatr 2025; 13:1567719. [PMID: 40171172 PMCID: PMC11958199 DOI: 10.3389/fped.2025.1567719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
The perinatal development period is critical for the formation of brain structures responsible for cognitive functions. Disruptions during this phase, such as perinatal asphyxia, characterized by impaired gas exchange and hypoxia, can lead to long-lasting neuronal damage and increased susceptibility to neurodegenerative diseases, including Alzheimer's disease (AD). AD, the most common cause of dementia globally, is marked by amyloid plaques, neurofibrillary tangles, and progressive cognitive decline. Emerging evidence links perinatal asphyxia with an elevated risk of AD, highlighting the potential role of oxidative stress, neuroinflammation, and epigenetic modifications as mediators. This review explores the mechanisms underlying brain damage after perinatal asphyxia, emphasizing oxidative stress, inflammation, and epigenetic changes that contribute to lifelong neurodegenerative susceptibility. Additionally, biomarkers identified in animal models reveal parallels between perinatal asphyxia and AD pathology, such as amyloid precursor protein alterations, gliosis, and microglial activation. These findings suggest perinatal asphyxia may prime microglia and epigenetically alter gene expression, predisposing individuals to chronic neurodegeneration. Future research should leverage advanced methodologies, including transcriptomics, epigenomics, and aged brain organoid models, to elucidate early-life influences on AD development. Understanding these mechanisms may pave the way for novel prevention strategies targeting early-life risk factors for neurodegenerative diseases.
Collapse
Affiliation(s)
- Bruna Petrucelli Arruda
- Neurohistology Laboratory, Center for Computation, Mathematics and Cognition, Federal University of ABC, Sao Bernardo do Campo, São Paulo, Brazil
| | - Pamela Pinheiro Martins
- Neurohistology Laboratory, Center for Computation, Mathematics and Cognition, Federal University of ABC, Sao Bernardo do Campo, São Paulo, Brazil
| | - Alexandre Hiroaki Kihara
- Neurogenetics Laboratory, Center for Computation, Mathematics and Cognition, Federal University of ABC, Sao Bernardo do Campo, São Paulo, Brazil
| | - Silvia Honda Takada
- Neurohistology Laboratory, Center for Computation, Mathematics and Cognition, Federal University of ABC, Sao Bernardo do Campo, São Paulo, Brazil
| |
Collapse
|
4
|
Hadzibegovic S, Bontempi B, Nicole O. Investigating the Impact of NMDA Receptor Organization and Biological Sex in the APPswe/PS1dE9 Mouse Model of Alzheimer's Disease. Int J Mol Sci 2025; 26:1737. [PMID: 40004200 PMCID: PMC11855313 DOI: 10.3390/ijms26041737] [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: 12/18/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by memory loss and cognitive decline, with women being disproportionately affected in both prevalence and severity. A key feature of AD is synaptic loss, particularly around amyloid-β (Aβ) aggregates, which correlates strongly with the severity of dementia. Oligomeric Aβ is believed to be the primary driver of synaptic dysfunction by impairing excitatory neurotransmission through interactions with synaptic receptors, including N-methyl-D-aspartate (NMDA) receptors. However, the influence of sex on these synaptic changes and NMDA receptor mislocalization in AD is not well understood. This study examined potential sex-specific differences in synaptotoxicity and the role of extrasynaptic GluN2B-containing NMDA receptors in AD pathogenesis using the APP/PS1 double transgenic mouse model. Although both male and female mice showed a similar amyloid burden and cognitive impairments, synaptic alterations were slightly less severe in females, suggesting subtle sex differences in synaptic pathology. Both sexes exhibited the mislocalization of GluN2B subunits to extrasynaptic areas, which was linked to reduced PSD-95 levels and the synaptic accumulation of Aβ1-42. Intrahippocampal injections of DL-TBOA confirmed the role of extrasynaptic GluN2B-containing NMDA receptors in memory dysfunction. These findings emphasize the importance of targeting synaptic receptor trafficking to address AD-related memory deficits, potentially offering a therapeutic approach for both sexes.
Collapse
Affiliation(s)
- Senka Hadzibegovic
- Neurocentre Magendie, INSERM U1215, 33077 Bordeaux, France;
- University of Bordeaux, 33077 Bordeaux, France;
| | - Bruno Bontempi
- University of Bordeaux, 33077 Bordeaux, France;
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine, CNRS UMR 5287, 33000 Bordeaux, France
| | - Olivier Nicole
- University of Bordeaux, 33077 Bordeaux, France;
- Institut Interdisciplinaire de Neurosciences, CNRS, UMR 5297, 33077 Bordeaux, France
| |
Collapse
|
5
|
Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, Chaudhary K, Sharma S, Sinha A, Abomughaid MM, Imran M, Lakhanpal S, Babalghith AO, Abu-Seer EA, Avinash D, Alzahrani HA, Alhindi AA, Iqbal D, Kumar S, Jha NK, Alghamdi S. Inflammatory signaling pathways in Alzheimer's disease: Mechanistic insights and possible therapeutic interventions. Ageing Res Rev 2025; 104:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [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: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The complex pathophysiology of Alzheimer's disease (AD) poses challenges for the development of therapies. Recently, neuroinflammation has been identified as a key pathogenic mechanism underlying AD, while inflammation has emerged as a possible target for the management and prevention of AD. Several prior studies have demonstrated that medications modulating neuroinflammation might lessen AD symptoms, mostly by controlling neuroinflammatory signaling pathways such as the NF-κB, MAPK, NLRP3, etc, and their respective signaling cascade. Moreover, targeting these inflammatory modalities with inhibitors, natural products, and metabolites has been the subject of intensive research because of their anti-inflammatory characteristics, with many studies demonstrating noteworthy pharmacological capabilities and potential clinical applications. Therefore, targeting inflammation is considered a promising strategy for treating AD. This review comprehensively elucidates the neuroinflammatory mechanisms underlying AD progression and the beneficial effects of inhibitors, natural products, and metabolites in AD treatment.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ashish Kumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Swati Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Eman Adnan Abu-Seer
- Department of Epidemiology and Medical Statistic, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, Saudi Arabia
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Hassan A Alzahrani
- Department of Respiratory Care, Medical Cities at the Minister of Interior, MCMOl, Riyadh, Saudi Arabia
| | | | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India; DST-FIST Laboratory, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology (SBT), Galgotias University, Greater Noida, India; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
6
|
Walters JM, Noblet HA, Chung HJ. An emerging role of STriatal-Enriched protein tyrosine Phosphatase in hyperexcitability-associated brain disorders. Neurobiol Dis 2024; 200:106641. [PMID: 39159894 DOI: 10.1016/j.nbd.2024.106641] [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: 04/27/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a brain-specific tyrosine phosphatase that is associated with numerous neurological and neuropsychiatric disorders. STEP dephosphorylates and inactivates various kinases and phosphatases critical for neuronal function and health including Fyn, Pyk2, ERK1/2, p38, and PTPα. Importantly, STEP dephosphorylates NMDA and AMPA receptors, two major glutamate receptors that mediate fast excitatory synaptic transmission. This STEP-mediated dephosphorylation leads to their internalization and inhibits both Hebbian synaptic potentiation and homeostatic synaptic scaling. Hence, STEP has been widely accepted to weaken excitatory synaptic strength. However, emerging evidence implicates a novel role of STEP in neuronal hyperexcitability and seizure disorders. Genetic deletion and pharmacological blockade of STEP reduces seizure susceptibility in acute seizure mouse models and audiogenic seizures in a mouse model of Fragile X syndrome. Pharmacologic inhibition of STEP also decreases hippocampal activity and neuronal intrinsic excitability. Here, we will highlight the divergent roles of STEP in excitatory synaptic transmission and neuronal intrinsic excitability, present the potential underlying mechanisms, and discuss their impact on STEP-associated neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jennifer M Walters
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hayden A Noblet
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
7
|
Nho K, Risacher SL, Apostolova LG, Bice PJ, Brosch JR, Deardorff R, Faber K, Farlow MR, Foroud T, Gao S, Rosewood T, Kim JP, Nudelman K, Yu M, Aisen P, Sperling R, Hooli B, Shcherbinin S, Svaldi D, Jack CR, Jagust WJ, Landau S, Vasanthakumar A, Waring JF, Doré V, Laws SM, Masters CL, Porter T, Rowe CC, Villemagne VL, Dumitrescu L, Hohman TJ, Libby JB, Mormino E, Buckley RF, Johnson K, Yang HS, Petersen RC, Ramanan VK, Ertekin-Taner N, Vemuri P, Cohen AD, Fan KH, Kamboh MI, Lopez OL, Bennett DA, Ali M, Benzinger T, Cruchaga C, Hobbs D, De Jager PL, Fujita M, Jadhav V, Lamb BT, Tsai AP, Castanho I, Mill J, Weiner MW, Saykin AJ. CYP1B1-RMDN2 Alzheimer's disease endophenotype locus identified for cerebral tau PET. Nat Commun 2024; 15:8251. [PMID: 39304655 PMCID: PMC11415491 DOI: 10.1038/s41467-024-52298-2] [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: 11/14/2023] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Determining the genetic architecture of Alzheimer's disease pathologies can enhance mechanistic understanding and inform precision medicine strategies. Here, we perform a genome-wide association study of cortical tau quantified by positron emission tomography in 3046 participants from 12 independent studies. The CYP1B1-RMDN2 locus is associated with tau deposition. The most significant signal is at rs2113389, explaining 4.3% of the variation in cortical tau, while APOE4 rs429358 accounts for 3.6%. rs2113389 is associated with higher tau and faster cognitive decline. Additive effects, but no interactions, are observed between rs2113389 and diagnosis, APOE4, and amyloid beta positivity. CYP1B1 expression is upregulated in AD. rs2113389 is associated with higher CYP1B1 expression and methylation levels. Mouse model studies provide additional functional evidence for a relationship between CYP1B1 and tau deposition but not amyloid beta. These results provide insight into the genetic basis of cerebral tau deposition and support novel pathways for therapeutic development in AD.
Collapse
Affiliation(s)
- Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of BioHealth Informatics, Indiana University, Indianapolis, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Liana G Apostolova
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Paula J Bice
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Jared R Brosch
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Rachael Deardorff
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Kelley Faber
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, USA
| | - Martin R Farlow
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA
| | - Tatiana Foroud
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, USA
| | - Thea Rosewood
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Jun Pyo Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Kelly Nudelman
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, USA
| | - Meichen Yu
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
| | - Paul Aisen
- Department of Neurology, Keck School of Medicine, University of Southern California, San Diego, USA
| | - Reisa Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | | | | | | | | - William J Jagust
- UC Berkeley Helen Wills Neuroscience Institute, University of California - Berkeley, Berkeley, USA
| | - Susan Landau
- UC Berkeley Helen Wills Neuroscience Institute, University of California - Berkeley, Berkeley, USA
| | | | | | - Vincent Doré
- CSIRO Health and Biosecurity, Melbourne, Australia
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Australia
| | - Simon M Laws
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Parkville, Australia
| | - Tenielle Porter
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Australia
- Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Parkville, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy, Austin Health, Heidelberg, Australia
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Logan Dumitrescu
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, USA
| | - Julia B Libby
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, USA
| | - Elizabeth Mormino
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, USA
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Keith Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Center for Alzheimer's Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | | | | | - Nilüfer Ertekin-Taner
- Department of Neurology, Mayo Clinic, Jacksonville, USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, USA
| | | | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Kang-Hsien Fan
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Oscar L Lopez
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Medical College, Rush University, Chicago, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University, St. Louis, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, USA
| | - Diana Hobbs
- Department of Radiology, Washington University School of Medicine, St. Louis, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, USA
| | - Vaishnavi Jadhav
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
| | - Bruce T Lamb
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
| | - Andy P Tsai
- Department of Neurology & Neurological Sciences, Stanford University, Stanford, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, USA
| | - Isabel Castanho
- Department for Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Jonathan Mill
- Department for Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Michael W Weiner
- Departments of Radiology, Medicine, and Psychiatry, University of California-San Francisco, San Francisco, USA
- Department of Veterans Affairs Medical Center, San Francisco, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, USA.
- Department of Neurology, Indiana University School of Medicine, Indianapolis, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA.
| |
Collapse
|
8
|
Ray A, Loghinov I, Ravindranath V, Barth AL. Early hippocampal hyperexcitability and synaptic reorganization in mouse models of amyloidosis. iScience 2024; 27:110629. [PMID: 39262788 PMCID: PMC11388185 DOI: 10.1016/j.isci.2024.110629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/09/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
The limited success of plaque-reducing therapies in Alzheimer's disease suggests that early treatment might be more effective in delaying or reversing memory impairments. Toward this end, it is important to establish the progression of synaptic and circuit changes before onset of plaques or cognitive deficits. Here, we used quantitative, fluorescence-based methods for synapse detection in CA1 pyramidal neurons to investigate the interaction between abnormal circuit activity, measured by Fos-immunoreactivity, and synapse reorganization in mouse models of amyloidosis. Using a genetically encoded, fluorescently labeled synaptic marker in juvenile mice (prior to sexual maturity), we find both synapse gain and loss depending on dendritic location. This progresses to broad synapse loss in aged mice. Elevated hippocampal activity in both CA3 and CA1 was present at weaning and preceded this reorganization. Thus, Aβ overproduction may initiate abnormal activity and subsequent input-specific synapse plasticity. These findings indicate that sustained amyloidosis drives heterogeneous and progressive circuit-wide abnormalities.
Collapse
Affiliation(s)
- Ajit Ray
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Iulia Loghinov
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Vijayalakshmi Ravindranath
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka 560012, India
- Centre for Brain Research, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Alison L. Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
9
|
Kuhn MK, Proctor EA. Microglial Drivers of Alzheimer's Disease Pathology: An Evolution of Diverse Participating States. Proteins 2024:10.1002/prot.26723. [PMID: 39219300 PMCID: PMC11871049 DOI: 10.1002/prot.26723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune-competent cells of the brain, become dysfunctional in Alzheimer's disease (AD), and their aberrant immune responses contribute to the accumulation of pathological proteins and neuronal injury. Genetic studies implicate microglia in the development of AD, prompting interest in developing immunomodulatory therapies to prevent or ameliorate disease. However, microglia take on diverse functional states in disease, playing both protective and detrimental roles in AD, which largely overlap and may shift over the disease course, complicating the identification of effective therapeutic targets. Extensive evidence gathered using transgenic mouse models supports an active role of microglia in pathology progression, though results vary and can be contradictory between different types of models and the degree of pathology at the time of study. Here, we review microglial immune signaling and responses that contribute to the accumulation and spread of pathological proteins or directly affect neuronal health. We additionally explore the use of induced pluripotent stem cell (iPSC)-derived models to study living human microglia and how they have contributed to our knowledge of AD and may begin to fill in the gaps left by mouse models. Ultimately, mouse and iPSC-derived models have their own limitations, and a comprehensive understanding of microglial dysfunction in AD will only be established by an integrated view across models and an appreciation for their complementary viewpoints and limitations.
Collapse
Affiliation(s)
- Madison K. Kuhn
- Department of Biomedical Engineering, Penn State University
- Department of Neurosurgery, Penn State College of Medicine
- Department of Pharmacology, Penn State College of Medicine
- Center for Neural Engineering, Penn State University
| | - Elizabeth A. Proctor
- Department of Biomedical Engineering, Penn State University
- Department of Neurosurgery, Penn State College of Medicine
- Department of Pharmacology, Penn State College of Medicine
- Center for Neural Engineering, Penn State University
- Department of Engineering Science & Mechanics, Penn State University
| |
Collapse
|
10
|
Pomilio C, Presa J, Oses C, Vinuesa A, Bentivegna M, Gregosa A, Riudavets M, Sevlever G, Galvan V, Levi V, Beauquis J, Saravia F. Loss of Direct Vascular Contact to Astrocytes in the Hippocampus as an Initial Event in Alzheimer's Disease. Evidence from Patients, In Vivo and In Vitro Experimental Models. Mol Neurobiol 2024; 61:5142-5160. [PMID: 38172288 DOI: 10.1007/s12035-023-03897-5] [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: 08/30/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of aggregated amyloid peptides in the brain parenchyma and within the walls of cerebral vessels. The hippocampus-a complex brain structure with a pivotal role in learning and memory-is implicated in this disease. However, there is limited data on vascular changes during AD pathological degeneration in this susceptible structure, which has distinctive vascular traits. Our aim was to evaluate vascular alterations in the hippocampus of AD patients and PDAPP-J20 mice-a model of AD-and to determine the impact of Aβ40 and Aβ42 on endothelial cell activation. We found a loss of physical astrocyte-endothelium interaction in the hippocampus of individuals with AD as compared to non-AD donors, along with reduced vascular density. Astrocyte-endothelial interactions and levels of the tight junction protein occludin were altered early in PDAPP-J20 mice, preceding any signs of morphological changes or disruption of the blood-brain barrier in these mice. At later stages, PDAPP-J20 mice exhibited decreased vascular density in the hippocampus and leakage of fluorescent tracers, indicating dysfunction of the vasculature and the BBB. In vitro studies showed that soluble Aβ40 exposure in human brain microvascular endothelial cells (HBMEC) was sufficient to induce NFκB translocation to the nucleus, which may be linked with an observed reduction in occludin levels. The inhibition of the membrane receptor for advanced glycation end products (RAGE) prevented these changes in HBMEC. Additional results suggest that Aβ42 indirectly affects the endothelium by inducing astrocytic factors. Furthermore, our results from human and mouse brain samples provide evidence for the crucial involvement of the hippocampal vasculature in Alzheimer's disease.
Collapse
Affiliation(s)
- C Pomilio
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - J Presa
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - C Oses
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - A Vinuesa
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - M Bentivegna
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - A Gregosa
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - M Riudavets
- FLENI, Instituto de Investigaciones Neurológicas Dr Raúl Carrea, Buenos Aires, Argentina
| | - G Sevlever
- FLENI, Instituto de Investigaciones Neurológicas Dr Raúl Carrea, Buenos Aires, Argentina
| | - V Galvan
- Department of Biochemistry and Molecular Biology and Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences, Oklahoma City, OK, 73104, USA
| | - V Levi
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - J Beauquis
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - F Saravia
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina.
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
11
|
Azargoonjahromi A. The duality of amyloid-β: its role in normal and Alzheimer's disease states. Mol Brain 2024; 17:44. [PMID: 39020435 PMCID: PMC11256416 DOI: 10.1186/s13041-024-01118-1] [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: 04/29/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024] Open
Abstract
Alzheimer's disease (AD) is a degenerative neurological condition that gradually impairs cognitive abilities, disrupts memory retention, and impedes daily functioning by impacting the cells of the brain. A key characteristic of AD is the accumulation of amyloid-beta (Aβ) plaques, which play pivotal roles in disease progression. These plaques initiate a cascade of events including neuroinflammation, synaptic dysfunction, tau pathology, oxidative stress, impaired protein clearance, mitochondrial dysfunction, and disrupted calcium homeostasis. Aβ accumulation is also closely associated with other hallmark features of AD, underscoring its significance. Aβ is generated through cleavage of the amyloid precursor protein (APP) and plays a dual role depending on its processing pathway. The non-amyloidogenic pathway reduces Aβ production and has neuroprotective and anti-inflammatory effects, whereas the amyloidogenic pathway leads to the production of Aβ peptides, including Aβ40 and Aβ42, which contribute to neurodegeneration and toxic effects in AD. Understanding the multifaceted role of Aβ, particularly in AD, is crucial for developing effective therapeutic strategies that target Aβ metabolism, aggregation, and clearance with the aim of mitigating the detrimental consequences of the disease. This review aims to explore the mechanisms and functions of Aβ under normal and abnormal conditions, particularly in AD, by examining both its beneficial and detrimental effects.
Collapse
|
12
|
Haynes JR, Whitmore CA, Behof WJ, Landman CA, Ong HH, Feld AP, Suero IC, Greer CB, Gore JC, Wijesinghe P, Matsubara JA, Wadzinski BE, Spiller BW, Pham W. Targeting soluble amyloid-beta oligomers with a novel nanobody. Sci Rep 2024; 14:16086. [PMID: 38992064 PMCID: PMC11239946 DOI: 10.1038/s41598-024-66970-6] [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: 02/09/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024] Open
Abstract
The classical amyloid cascade hypothesis postulates that the aggregation of amyloid plaques and the accumulation of intracellular hyperphosphorylated Tau tangles, together, lead to profound neuronal death. However, emerging research has demonstrated that soluble amyloid-β oligomers (SAβOs) accumulate early, prior to amyloid plaque formation. SAβOs induce memory impairment and disrupt cognitive function independent of amyloid-β plaques, and even in the absence of plaque formation. This work describes the development and characterization of a novel anti-SAβO (E3) nanobody generated from an alpaca immunized with SAβO. In-vitro assays and in-vivo studies using 5XFAD mice indicate that the fluorescein (FAM)-labeled E3 nanobody recognizes both SAβOs and amyloid-β plaques. The E3 nanobody traverses across the blood-brain barrier and binds to amyloid species in the brain of 5XFAD mice. Imaging of mouse brains reveals that SAβO and amyloid-β plaques are not only different in size, shape, and morphology, but also have a distinct spatial distribution in the brain. SAβOs are associated with neurons, while amyloid plaques reside in the extracellular matrix. The results of this study demonstrate that the SAβO nanobody can serve as a diagnostic agent with potential theragnostic applications in Alzheimer's disease.
Collapse
Affiliation(s)
- Justin R Haynes
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Clayton A Whitmore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - William J Behof
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Charlotte A Landman
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Henry H Ong
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Andrew P Feld
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Isabelle C Suero
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Celeste B Greer
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
- Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Printha Wijesinghe
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, V5Z3N9, Canada
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC, V5Z3N9, Canada
| | - Brian E Wadzinski
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA.
| | - Benjamin W Spiller
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Center for Structural Biology, Vanderbilt University, Nashville, TN, 37235, USA.
| | - Wellington Pham
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA.
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
- Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA.
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, 37212, USA.
| |
Collapse
|
13
|
Żukowska J, Moss SJ, Subramanian V, Acharya KR. Molecular basis of selective amyloid-β degrading enzymes in Alzheimer's disease. FEBS J 2024; 291:2999-3029. [PMID: 37622248 DOI: 10.1111/febs.16939] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/31/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023]
Abstract
The accumulation of the small 42-residue long peptide amyloid-β (Aβ) has been proposed as a major trigger for the development of Alzheimer's disease (AD). Within the brain, the concentration of Aβ peptide is tightly controlled through production and clearance mechanisms. Substantial experimental evidence now shows that reduced levels of Aβ clearance are present in individuals living with AD. This accumulation of Aβ can lead to the formation of large aggregated amyloid plaques-one of two detectable hallmarks of the disease. Aβ-degrading enzymes (ADEs) are major players in the clearance of Aβ. Stimulating ADE activity or expression, in order to compensate for the decreased clearance in the AD phenotype, provides a promising therapeutic target. It has been reported in mice that upregulation of ADEs can reduce the levels of Aβ peptide and amyloid plaques-in some cases, this led to improved cognitive function. Among several known ADEs, neprilysin (NEP), endothelin-converting enzyme-1 (ECE-1), insulin degrading enzyme (IDE) and angiotensin-1 converting enzyme (ACE) from the zinc metalloprotease family have been identified as important. These ADEs have the capacity to digest soluble Aβ which, in turn, cannot form the toxic oligomeric species. While they are known for their amyloid degradation, they exhibit complexity through promiscuous nature and a broad range of substrates that they can degrade. This review highlights current structural and functional understanding of these key ADEs, giving some insight into the molecular interactions that leads to the hydrolysis of peptide substrates, the crucial tasks performed by them and the potential for therapeutic use in the future.
Collapse
|
14
|
Collins HM, Greenfield S. Rodent Models of Alzheimer's Disease: Past Misconceptions and Future Prospects. Int J Mol Sci 2024; 25:6222. [PMID: 38892408 PMCID: PMC11172947 DOI: 10.3390/ijms25116222] [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: 04/03/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with no effective treatments, not least due to the lack of authentic animal models. Typically, rodent models recapitulate the effects but not causes of AD, such as cholinergic neuron loss: lesioning of cholinergic neurons mimics the cognitive decline reminiscent of AD but not its neuropathology. Alternative models rely on the overexpression of genes associated with familial AD, such as amyloid precursor protein, or have genetically amplified expression of mutant tau. Yet transgenic rodent models poorly replicate the neuropathogenesis and protein overexpression patterns of sporadic AD. Seeding rodents with amyloid or tau facilitates the formation of these pathologies but cannot account for their initial accumulation. Intracerebral infusion of proinflammatory agents offer an alternative model, but these fail to replicate the cause of AD. A novel model is therefore needed, perhaps similar to those used for Parkinson's disease, namely adult wildtype rodents with neuron-specific (dopaminergic) lesions within the same vulnerable brainstem nuclei, 'the isodendritic core', which are the first to degenerate in AD. Site-selective targeting of these nuclei in adult rodents may recapitulate the initial neurodegenerative processes in AD to faithfully mimic its pathogenesis and progression, ultimately leading to presymptomatic biomarkers and preventative therapies.
Collapse
Affiliation(s)
- Helen M. Collins
- Neuro-Bio Ltd., Building F5 The Culham Campus, Abingdon OX14 3DB, UK;
| | | |
Collapse
|
15
|
Dashkova AS, Kovalev VI, Chaplygina AV, Zhdanova DY, Bobkova NV. Unique Properties of Synaptosomes and Prospects for Their Use for the Treatment of Alzheimer's Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1031-1044. [PMID: 38981699 DOI: 10.1134/s0006297924060051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative condition affecting millions worldwide. Prevalence of AD correlates with increased life expectancy and aging population in the developed countries. Considering that AD is a multifactorial disease involving various pathological processes such as synaptic dysfunction, neuroinflammation, oxidative stress, and improper protein folding, a comprehensive approach targeting multiple pathways may prove effective in slowing the disease progression. Cellular therapy and its further development in the form of cell vesicle and particularly mitochondrial transplantation represent promising approaches for treating neurodegeneration. The use of synaptosomes, due to uniqueness of their contents, could mark a new stage in the development of comprehensive therapies for neurodegenerative diseases, particularly AD. Synaptosomes contain unique memory mitochondria, which differ not only in size but also in functionality compared to the mitochondria in the neuronal soma. These synaptosomal mitochondria actively participate in cellular communication and signal transmission within synapses. Synaptosomes also contain other elements such as their own protein synthesis machinery, synaptic vesicles with neurotransmitters, synaptic adhesion molecules, and microRNAs - all crucial for synaptic transmission and, consequently, cognitive processes. Complex molecular ensemble ensures maintenance of the synaptic autonomy of mitochondria. Additionally, synaptosomes, with their affinity for neurons, can serve as an optimal platform for targeted drug delivery to nerve cells. This review discusses unique composition of synaptosomes, their capabilities and advantages, as well as limitations of their suggested use as therapeutic agents for treating neurodegenerative pathologies, particularly AD.
Collapse
Affiliation(s)
- Alla S Dashkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vladimir I Kovalev
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Alina V Chaplygina
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Daria Yu Zhdanova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Natalia V Bobkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
16
|
Spoleti E, La Barbera L, Cauzzi E, De Paolis ML, Saba L, Marino R, Sciamanna G, Di Lazzaro V, Keller F, Nobili A, Krashia P, D'Amelio M. Dopamine neuron degeneration in the Ventral Tegmental Area causes hippocampal hyperexcitability in experimental Alzheimer's Disease. Mol Psychiatry 2024; 29:1265-1280. [PMID: 38228889 PMCID: PMC11189820 DOI: 10.1038/s41380-024-02408-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/17/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Abstract
Early and progressive dysfunctions of the dopaminergic system from the Ventral Tegmental Area (VTA) have been described in Alzheimer's Disease (AD). During the long pre-symptomatic phase, alterations in the function of Parvalbumin interneurons (PV-INs) are also observed, resulting in cortical hyperexcitability represented by subclinical epilepsy and aberrant gamma-oscillations. However, it is unknown whether the dopaminergic deficits contribute to brain hyperexcitability in AD. Here, using the Tg2576 mouse model of AD, we prove that reduced hippocampal dopaminergic innervation, due to VTA dopamine neuron degeneration, impairs PV-IN firing and gamma-waves, weakens the inhibition of pyramidal neurons and induces hippocampal hyperexcitability via lower D2-receptor-mediated activation of the CREB-pathway. These alterations coincide with reduced PV-IN numbers and Perineuronal Net density. Importantly, L-DOPA and the selective D2-receptor agonist quinpirole rescue p-CREB levels and improve the PV-IN-mediated inhibition, thus reducing hyperexcitability. Moreover, similarly to quinpirole, sumanirole - another D2-receptor agonist and a known anticonvulsant - not only increases p-CREB levels in PV-INs but also restores gamma-oscillations in Tg2576 mice. Conversely, blocking the dopaminergic transmission with sulpiride (a D2-like receptor antagonist) in WT mice reduces p-CREB levels in PV-INs, mimicking what occurs in Tg2576. Overall, these findings support the hypothesis that the VTA dopaminergic system integrity plays a key role in hippocampal PV-IN function and survival, disclosing a relevant contribution of the reduced dopaminergic tone to aberrant gamma-waves, hippocampal hyperexcitability and epileptiform activity in early AD.
Collapse
Affiliation(s)
- Elena Spoleti
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Livia La Barbera
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Emma Cauzzi
- Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Maria Luisa De Paolis
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Luana Saba
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Ramona Marino
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Giuseppe Sciamanna
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
- UniCamillus International University of Health Sciences, 00131, Rome, Italy
| | - Vincenzo Di Lazzaro
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
- Operative Research Unit of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128, Rome, Italy
| | - Flavio Keller
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Paraskevi Krashia
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Marcello D'Amelio
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy.
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.
| |
Collapse
|
17
|
Chockanathan U, Padmanabhan K. Differential disruptions in population coding along the dorsal-ventral axis of CA1 in the APP/PS1 mouse model of Aβ pathology. PLoS Comput Biol 2024; 20:e1012085. [PMID: 38709845 PMCID: PMC11098488 DOI: 10.1371/journal.pcbi.1012085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/16/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024] Open
Abstract
Alzheimer's Disease (AD) is characterized by a range of behavioral alterations, including memory loss and psychiatric symptoms. While there is evidence that molecular pathologies, such as amyloid beta (Aβ), contribute to AD, it remains unclear how this histopathology gives rise to such disparate behavioral deficits. One hypothesis is that Aβ exerts differential effects on neuronal circuits across brain regions, depending on the neurophysiology and connectivity of different areas. To test this, we recorded from large neuronal populations in dorsal CA1 (dCA1) and ventral CA1 (vCA1), two hippocampal areas known to be structurally and functionally diverse, in the APP/PS1 mouse model of amyloidosis. Despite similar levels of Aβ pathology, dCA1 and vCA1 showed distinct disruptions in neuronal population activity as animals navigated a virtual reality environment. In dCA1, pairwise correlations and entropy, a measure of the diversity of activity patterns, were decreased in APP/PS1 mice relative to age-matched C57BL/6 controls. However, in vCA1, APP/PS1 mice had increased pair-wise correlations and entropy as compared to age matched controls. Finally, using maximum entropy models, we connected the microscopic features of population activity (correlations) to the macroscopic features of the population code (entropy). We found that the models' performance increased in predicting dCA1 activity, but decreased in predicting vCA1 activity, in APP/PS1 mice relative to the controls. Taken together, we found that Aβ exerts distinct effects across different hippocampal regions, suggesting that the various behavioral deficits of AD may reflect underlying heterogeneities in neuronal circuits and the different disruptions that Aβ pathology causes in those circuits.
Collapse
Affiliation(s)
- Udaysankar Chockanathan
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Neuroscience Graduate Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Krishnan Padmanabhan
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Neuroscience Graduate Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Center for Visual Sciences, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Intellectual and Developmental Disabilities Research Center, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| |
Collapse
|
18
|
Fong V, Kanuri B, Traubert O, Lui M, Patel SB. Behavioral and Metabolic Effects of ABCG4 KO in the APP swe,Ind (J9) Mouse Model of Alzheimer's Disease. J Mol Neurosci 2024; 74:49. [PMID: 38668787 PMCID: PMC11052713 DOI: 10.1007/s12031-024-02214-6] [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: 06/02/2023] [Accepted: 03/21/2024] [Indexed: 04/29/2024]
Abstract
The pathogenesis of Alzheimer's disease (AD) is complex and involves an imbalance between production and clearance of amyloid-ß peptides (Aß), resulting in accumulation of Aß in senile plaques. Hypercholesterolemia is a major risk factor for developing AD, with cholesterol shown to accumulate in senile plaques and increase production of Aß. ABCG4 is a member of the ATP-binding cassette transporters predominantly expressed in the CNS and has been suggested to play a role in cholesterol and Aß efflux from the brain. In this study, we bred Abcg4 knockout (KO) with the APPSwe,Ind (J9) mouse model of AD to test the hypothesis that loss of Abcg4 would exacerbate the AD phenotype. Unexpectedly, no differences were observed in novel object recognition (NOR) and novel object placement (NOP) behavioral tests, or on histologic examinations of brain tissues for senile plaque numbers. Furthermore, clearance of radiolabeled Aß from the brains did not differ between Abcg4 KO and control mice. Metabolic testing by indirect calorimetry, glucose tolerance test (GTT), and insulin tolerance test (ITT) were also mostly similar between groups with only a few mild metabolic differences noted. Overall, these data suggest that the loss of ABCG4 did not exacerbate the AD phenotype.
Collapse
Affiliation(s)
- Vincent Fong
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati, OH, USA
| | - Babunageswararao Kanuri
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati, OH, USA
| | - Owen Traubert
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati, OH, USA
| | - Min Lui
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Shailendra B Patel
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
19
|
Chen A, Shea D, Daggett V. Performance of SOBA-AD blood test in discriminating Alzheimer's disease patients from cognitively unimpaired controls in two independent cohorts. Sci Rep 2024; 14:7946. [PMID: 38575622 PMCID: PMC10995183 DOI: 10.1038/s41598-024-57107-w] [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: 01/02/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
Amyloid-beta (Aβ) toxic oligomers are critical early players in the molecular pathology of Alzheimer's disease (AD). We have developed a Soluble Oligomer Binding Assay (SOBA-AD) for detection of these Aβ oligomers that contain α-sheet secondary structure that discriminates plasma samples from patients on the AD continuum from non-AD controls. We tested 265 plasma samples from two independent cohorts to investigate the performance of SOBA-AD. Testing was performed at two different sites, with different personnel, reagents, and instrumentation. Across two cohorts, SOBA-AD discriminated AD patients from cognitively unimpaired (CU) subjects with 100% sensitivity, > 95% specificity, and > 98% area under the curve (AUC) (95% CI 0.95-1.00). A SOBA-AD positive readout, reflecting α-sheet toxic oligomer burden, was found in AD patients, and not in controls, providing separation of the two populations, aside from 5 SOBA-AD positive controls. Based on an earlier SOBA-AD study, the Aβ oligomers detected in these CU subjects may represent preclinical cases of AD. The results presented here support the value of SOBA-AD as a promising blood-based tool for the detection and confirmation of AD.
Collapse
Affiliation(s)
- Amy Chen
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA
| | - Dylan Shea
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA
- University of Washington, Box 355610, Seattle, WA, 98195-5610, USA
| | - Valerie Daggett
- AltPep Corporation, 1150 Eastlake Avenue N, Suite 800, Seattle, WA, 98109, USA.
- University of Washington, Box 355610, Seattle, WA, 98195-5610, USA.
| |
Collapse
|
20
|
Kuhn MK, Kang RY, Kim C, Tagay Y, Morris N, Tabdanov ED, Elcheva IA, Proctor EA. Dynamic neuroinflammatory profiles predict Alzheimer's disease pathology in microglia-containing cerebral organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.16.567220. [PMID: 38014053 PMCID: PMC10680718 DOI: 10.1101/2023.11.16.567220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Neuroinflammation and the underlying dysregulated immune responses of microglia actively contribute to the progression and, likely, the initiation of Alzheimer's disease (AD). Fine-tuned therapeutic modulation of immune dysfunction to ameliorate disease cannot be achieved without the characterization of diverse microglial states that initiate unique, and sometimes contradictory, immune responses that evolve over time in chronic inflammatory environments. Because of the functional differences between human and murine microglia, untangling distinct, disease-relevant reactive states and their corresponding effects on pathology or neuronal health may not be possible without the use of human cells. In order to profile shifting microglial states in early AD and identify microglia-specific drivers of disease, we differentiated human induced pluripotent stem cells (iPSCs) carrying a familial AD PSEN2 mutation or its isogenic control into cerebral organoids and quantified the changes in cytokine concentrations over time with Luminex XMAP technology. We used partial least squares (PLS) modeling to build cytokine signatures predictive of disease and age to identify key differential patterns of cytokine expression that inform the overall organoid immune milieu and quantified the corresponding changes in protein pathology. AD organoids exhibited an overall reduction in cytokine secretion after an initial amplified immune response. We demonstrate that reduced synapse density observed in the AD organoids is prevented with microglial depletion. Crucially, these differential effects of dysregulated immune signaling occurred without the accumulation of pathological proteins. In this study, we used microglia-containing AD organoids to quantitatively characterize an evolving immune milieu, made up of a diverse of collection of activation patterns and immune responses, to identify how a dynamic, overall neuroinflammatory state negatively impacts neuronal health and the cell-specific contribution of microglia.
Collapse
Affiliation(s)
- Madison K Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Rachel Y Kang
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - ChaeMin Kim
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Yerbol Tagay
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Nathan Morris
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Erdem D Tabdanov
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
| | - Irina A Elcheva
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
21
|
Wang M, He X, Li J, Han D, You P, Yu H, Wang L, Su B. GDI2 deletion alleviates neurodegeneration and memory loss in the 5xFAD mice model of Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167093. [PMID: 38382624 DOI: 10.1016/j.bbadis.2024.167093] [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: 11/15/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/23/2024]
Abstract
Accumulation of insoluble deposits of amyloid β-peptide (Aβ), derived from amyloid precursor protein (APP) processing, represents one of the major pathological hallmarks of Alzheimer's disease (AD). Perturbations in APP transport and hydrolysis could lead to increased Aβ production. However, the precise mechanisms underlying APP transport remain elusive. The GDP dissociation inhibitor2 (GDI2), a crucial regulator of Rab GTPase activity and intracellular vesicle and membrane trafficking, was investigated for its impact on AD pathogenesis through neuron-specific knockout of GDI2 in 5xFAD mice. Notably, deficiency of GDI2 significantly ameliorated cognitive impairment, prevented neuronal loss in the subiculum and cortical layer V, reduced senile plaques as well as astrocyte activation in 5xFAD mice. Conversely, increased activated microglia and phagocytosis were observed in GDI2 ko mice. Further investigation revealed that GDI2 knockout led to more APP co-localized with the ER rather than the Golgi apparatus and endosomes in SH-SY5Y cells, resulting in decreased Aβ production. Collectively, these findings suggest that GDI2 may regulate Aβ production by modulating APP intracellular transport and localization dynamics. In summary, our study identifies GDI2 as a pivotal regulator governing APP transport and process implicated in AD pathology; thus highlighting its potential as an attractive pharmacological target for future drug development against AD.
Collapse
Affiliation(s)
- Meitian Wang
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiuqing He
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jie Li
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Daobin Han
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Pan You
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Hui Yu
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Luwen Wang
- Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Bo Su
- Department of Cell Biology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
22
|
Chan DC, Kim C, Kang RY, Kuhn MK, Beidler LM, Zhang N, Proctor EA. Cytokine expression patterns predict suppression of vulnerable neural circuits in a mouse model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.17.585383. [PMID: 38559177 PMCID: PMC10979954 DOI: 10.1101/2024.03.17.585383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease is a neurodegenerative disorder characterized by progressive amyloid plaque accumulation, tau tangle formation, neuroimmune dysregulation, synapse an neuron loss, and changes in neural circuit activation that lead to cognitive decline and dementia. Early molecular and cellular disease-instigating events occur 20 or more years prior to presentation of symptoms, making them difficult to study, and for many years amyloid-β, the aggregating peptide seeding amyloid plaques, was thought to be the toxic factor responsible for cognitive deficit. However, strategies targeting amyloid-β aggregation and deposition have largely failed to produce safe and effective therapies, and amyloid plaque levels poorly correlate with cognitive outcomes. However, a role still exists for amyloid-β in the variation in an individual's immune response to early, soluble forms of aggregates, and the downstream consequences of this immune response for aberrant cellular behaviors and creation of a detrimental tissue environment that harms neuron health and causes changes in neural circuit activation. Here, we perform functional magnetic resonance imaging of awake, unanesthetized Alzheimer's disease mice to map changes in functional connectivity over the course of disease progression, in comparison to wild-type littermates. In these same individual animals, we spatiotemporally profile the immune milieu by measuring cytokines, chemokines, and growth factors across various brain regions and over the course of disease progression from pre-pathology through established cognitive deficit. We identify specific signatures of immune activation predicting hyperactivity followed by suppression of intra- and then inter-regional functional connectivity in multiple disease-relevant brain regions, following the pattern of spread of amyloid pathology.
Collapse
Affiliation(s)
- Dennis C Chan
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neurotechnology in Mental Health Research, Pennsylvania State University, University Park, PA, USA
| | - ChaeMin Kim
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Rachel Y Kang
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Madison K Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Lynne M Beidler
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Nanyin Zhang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neurotechnology in Mental Health Research, Pennsylvania State University, University Park, PA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
23
|
Haynes JR, Whitmore CA, Behof WJ, Landman CA, Ong HH, Feld AP, Suero IC, Greer CB, Gore JC, Wijesinghe P, Matsubara JA, Wadzinski BE, Spiller BW, Pham W. TARGETING SOLUBLE AMYLOID-BETA OLIGOMERS WITH A NOVEL NANOBODY. RESEARCH SQUARE 2024:rs.3.rs-3944211. [PMID: 38559050 PMCID: PMC10980145 DOI: 10.21203/rs.3.rs-3944211/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The classical amyloid cascade hypothesis postulates that the aggregation of amyloid plaques and the accumulation of intracellular hyperphosphorylated Tau tangles, together, lead to profound neuronal death. However, emerging research has demonstrated that soluble amyloid-β oligomers (SAβOs) accumulate early, prior to amyloid plaque formation. SAβOs induce memory impairment and disrupt cognitive function independent of amyloid-β plaques, and even in the absence of plaque formation. This work describes the development and characterization of a novel anti-SAβO (E3) nanobody generated from an alpaca immunized with SAβO. In-vitro assays and in-vivo studies using 5XFAD mice indicate that the fluorescein (FAM)-labeled E3 nanobody recognizes both SAβOs and amyloid-β plaques. The E3 nanobody traverses across the blood-brain barrier and binds to amyloid species in the brain of 5XFAD mice. Imaging of mouse brains reveals that SAβO and amyloid-β plaques are not only different in size, shape, and morphology, but also have a distinct spatial distribution in the brain. SAβOs are associated with neurons, while amyloid plaques reside in the extracellular matrix. The results of this study demonstrate that the SAβO nanobody can serve as a diagnostic agent with potential theragnostic applications in Alzheimer's disease.
Collapse
Affiliation(s)
- Justin R. Haynes
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Clayton A. Whitmore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - William J. Behof
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Charlotte A. Landman
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Henry H. Ong
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrew P. Feld
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Isabelle C. Suero
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Celeste B. Greer
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - John C. Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Printha Wijesinghe
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, V5Z3N9, Canada
| | - Joanne A. Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, V5Z3N9, Canada
| | - Brian E. Wadzinski
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Benjamin W. Spiller
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Wellington Pham
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
24
|
Ali M, Wani SUD, Dey T, Sridhar SB, Qadrie ZL. A common molecular and cellular pathway in developing Alzheimer and cancer. Biochem Biophys Rep 2024; 37:101625. [PMID: 38225990 PMCID: PMC10788207 DOI: 10.1016/j.bbrep.2023.101625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024] Open
Abstract
Globally cancer and Alzheimer's disease (AD) are two major diseases and still, there is no clearly defined molecular mechanism. There is an opposite relation between cancer and AD which are the proportion of emerging cancer was importantly slower in AD patients, whereas slow emerging AD in patients with cancer. In cancer, regulation of cell mechanisms is interrupted by an increase in cell survival and proliferation, while on the contrary, AD is related to augmented neuronal death, that may be either produced by or associated with amyloid-β (Aβ) and tau deposition. Stated that the probability that disruption of mechanisms takes part in the regulation of cell survival/death and might be implicated in both diseases. The mechanism of actions such as DNA-methylation, genetic polymorphisms, or another mechanism of actions that induce alteration in the action of drugs with significant roles in resolving the finding to repair and live or die might take part in the pathogenesis of these two ailments. The functions of miRNA, p53, Pin1, the Wnt signaling pathway, PI3 KINASE/Akt/mTOR signaling pathway GRK2 signaling pathway, and the pathophysiological role of oxidative stress are presented in this review as potential candidates which hypothetically describe inverse relations between cancer and AD. Innovative materials almost mutual mechanisms in the aetiology of cancer and AD advocates novel treatment approaches. Among these treatment strategies, the most promising use treatment such as tyrosine kinase inhibitor, nilotinib, protein kinase C, and bexarotene.
Collapse
Affiliation(s)
- Mohammad Ali
- Department of Pharmacology, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B.G Nagar, Nagamagala, Bellur, Karnataka, 571418, India
- Department of Pharmacy Practice, East Point College of Pharmacy, Bangalore, 560049, India
| | - Shahid Ud Din Wani
- Division of Pharmaceutics, Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Srinagar, 190006, India
| | - Tathagata Dey
- Department of Pharmaceutical Chemistry, East Point College of Pharmacy, Bangalore, 560049, India
| | - Sathvik B. Sridhar
- Department of Clinical Pharmacy and Pharmacology, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, PO Box 11172, United Arab Emirates
| | | |
Collapse
|
25
|
Kim NY, Lee HY, Choi YY, Mo SJ, Jeon S, Ha JH, Park SD, Shim JJ, Lee J, Chung BG. Effect of gut microbiota-derived metabolites and extracellular vesicles on neurodegenerative disease in a gut-brain axis chip. NANO CONVERGENCE 2024; 11:7. [PMID: 38340254 DOI: 10.1186/s40580-024-00413-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/21/2024] [Indexed: 02/12/2024]
Abstract
A new perspective suggests that a dynamic bidirectional communication system, often referred to as the microbiome-gut-brain axis, exists among the gut, its microbiome, and the central nervous system (CNS). This system may influence brain health and various brain-related diseases, especially in the realms of neurodevelopmental and neurodegenerative conditions. However, the exact mechanism is not yet understood. Metabolites or extracellular vesicles derived from microbes in the gut have the capacity to traverse the intestinal epithelial barrier or blood-brain barrier, gaining access to the systemic circulation. This phenomenon can initiate the physiological responses that directly or indirectly impact the CNS and its function. However, reliable and controllable tools are required to demonstrate the causal effects of gut microbial-derived substances on neurogenesis and neurodegenerative diseases. The integration of microfluidics enhances scientific research by providing advanced in vitro engineering models. In this study, we investigated the impact of microbe-derived metabolites and exosomes on neurodevelopment and neurodegenerative disorders using human induced pluripotent stem cells (iPSCs)-derived neurons in a gut-brain axis chip. While strain-specific, our findings indicate that both microbial-derived metabolites and exosomes exert the significant effects on neural growth, maturation, and synaptic plasticity. Therefore, our results suggest that metabolites and exosomes derived from microbes hold promise as potential candidates and strategies for addressing neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Na Yeon Kim
- Department of Biomedical Engineering, Sogang University, Seoul, Korea
| | - Ho Yeon Lee
- Department of Biomedical Engineering, Sogang University, Seoul, Korea
| | - Yoon Young Choi
- Institute of Integrated Biotechnology, Sogang University, Seoul, Korea
| | | | | | - Jang Ho Ha
- Department of Mechanical Engineering, Sogang University, Seoul, Korea
| | | | | | | | - Bong Geun Chung
- Department of Biomedical Engineering, Sogang University, Seoul, Korea.
- Institute of Integrated Biotechnology, Sogang University, Seoul, Korea.
- Department of Mechanical Engineering, Sogang University, Seoul, Korea.
- Institute of Smart Biosensor, Sogang University, Seoul, Korea.
| |
Collapse
|
26
|
Català-Solsona J, Lutzu S, Lituma PJ, Fábregas-Ordoñez C, Siedlecki D, Giménez-Llort L, Miñano-Molina AJ, Saura CA, Castillo PE, Rodriguez-Álvarez J. Nr4a2 blocks oAβ-mediated synaptic plasticity dysfunction and ameliorates spatial memory deficits in the APP Sw,Ind mouse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.577010. [PMID: 38328087 PMCID: PMC10849715 DOI: 10.1101/2024.01.24.577010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Alzheimer's disease AD is associated with disruptions in neuronal communication, especially in brain regions crucial for learning and memory, such as the hippocampus. The amyloid hypothesis suggests that the accumulation of amyloid-beta oligomers (oAβ) contributes to synaptic dysfunction by internalisation of synaptic AMPA receptors. Recently, it has been reported that Nr4a2, a member of the Nr4a family of orphan nuclear receptors, plays a role in hippocampal synaptic plasticity by regulating BDNF and synaptic AMPA receptors. Here, we demonstrate that oAβ inhibits activity-dependent Nr4a2 activation in hippocampal neurons, indicating a potential link between oAβ and Nr4a2 down-regulation. Furthermore, we have observed a reduction in Nr4a2 protein levels in postmortem hippocampal tissue samples from early AD stages. Pharmacological activation of Nr4a2 proves effective in preventing oAβ-mediated synaptic depression in the hippocampus. Notably, Nr4a2 overexpression in the hippocampus of AD mouse models ameliorates spatial learning and memory deficits. In conclusion, the findings suggest that oAβ may contribute to early cognitive impairment in AD by blocking Nr4a2 activation, leading to synaptic dysfunction. Thus, our results further support that Nr4a2 activation is a potential therapeutic target to mitigate oAβ-induced synaptic and cognitive impairments in the early stages of Alzheimer's disease.
Collapse
|
27
|
Viana da Silva S, Haberl MG, Gaur K, Patel R, Narayan G, Ledakis M, Fu ML, de Castro Vieira M, Koo EH, Leutgeb JK, Leutgeb S. Localized APP expression results in progressive network dysfunction by disorganizing spike timing. Neuron 2024; 112:124-140.e6. [PMID: 37909036 PMCID: PMC10877582 DOI: 10.1016/j.neuron.2023.10.001] [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: 12/26/2022] [Revised: 06/16/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023]
Abstract
Progressive cognitive decline in Alzheimer's disease could either be caused by a spreading molecular pathology or by an initially focal pathology that causes aberrant neuronal activity in a larger network. To distinguish between these possibilities, we generated a mouse model with expression of mutant human amyloid precursor protein (APP) in only hippocampal CA3 cells. We found that performance in a hippocampus-dependent memory task was impaired in young adult and aged mutant mice. In both age groups, we then recorded from the CA1 region, which receives inputs from APP-expressing CA3 cells. We observed that theta oscillation frequency in CA1 was reduced along with disrupted relative timing of principal cells. Highly localized pathology limited to the presynaptic CA3 cells is thus sufficient to cause aberrant firing patterns in postsynaptic neuronal networks, which indicates that disease progression is not only from spreading pathology but also mediated by progressively advancing physiological dysfunction.
Collapse
Affiliation(s)
- Silvia Viana da Silva
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA; NeuroCure Excellence Cluster and German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Matthias G Haberl
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Neuroscience Research Center, Charitéplatz 1, 10117 Berlin, Germany
| | - Kshitij Gaur
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Rina Patel
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Gautam Narayan
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Max Ledakis
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Maylin L Fu
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Miguel de Castro Vieira
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Neuroscience Research Center, Charitéplatz 1, 10117 Berlin, Germany
| | - Edward H Koo
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jill K Leutgeb
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| | - Stefan Leutgeb
- Neurobiology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA; Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
28
|
Akbari S, Haghani M, Ghobadi M, Hooshmandi E, Haghighi AB, Salehi MS, Pandamooz S, Azarpira N, Afshari A, Zabihi S, Nemati M, Bayat M. Combination Therapy with Platelet-Rich Plasma and Epidermal Neural Crest Stem Cells Increases Treatment Efficacy in Vascular Dementia. Stem Cells Int 2023; 2023:3784843. [PMID: 38146481 PMCID: PMC10749736 DOI: 10.1155/2023/3784843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/07/2023] [Accepted: 11/28/2023] [Indexed: 12/27/2023] Open
Abstract
This study aimed to evaluate the efficacy and treatment mechanism of platelet-rich plasma (PRP) and neural crest-derived epidermal stem cells (ESCs) in their administration alone and combination in vascular dementia (VaD) model by two-vessel occlusion (2VO). Methods. Sixty-six rats were divided into six groups: the control, sham, 2VO + vehicle, 2VO + PRP, 2VO + ESC, and 2VO + ESC + PRP. The treated groups received 1 million cells on days 4, 14, and 21 with or without 500 µl PRP (twice a week) after 2VO. The memory performance and anxiety were evaluated by behavioral tests including open field, passive avoidance, and Morris water maze. The basal-synaptic transmission (BST) and long-term potentiation (LTP) were assessed through field-potential recordings of the CA1. The mRNA expression levels of IGF-1, TGF-β1, PSD-95, and GSk-3β were measured in the rat hippocampus by quantitative reverse transcription polymerase chain reaction. Results. The results demonstrated impaired learning, memory, and synaptic plasticity in the 2VO rats, along with a significant decrease in the expression of IGF-1, TGF-β1, PSD-95, and upregulation of GSK-3β. Treatment with ESC alone and ESC + PRP showed similar improvements in spatial memory and LTP induction, with associated upregulation of PSD-95 and downregulation of GSK-3β. However, only the ESC + PRP group showed recovery in BST. Furthermore, combination therapy was more effective than PRP monotherapy for LTP and memory. Conclusions. The transplantation of ESC showed better effects than PRP alone, and combination therapy increased the treatment efficacy with the recovery of BST. This finding may be a clue for the combination therapy of ESC and PRP for VaD.
Collapse
Affiliation(s)
- Somayeh Akbari
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Haghani
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Ghobadi
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Saied Salehi
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Shiraz Institute of Stem Cell and Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afsoon Afshari
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahrbanoo Zabihi
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Nemati
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
29
|
Li X, Quan M, Wei Y, Wang W, Xu L, Wang Q, Jia J. Critical thinking of Alzheimer's transgenic mouse model: current research and future perspective. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2711-2754. [PMID: 37480469 DOI: 10.1007/s11427-022-2357-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/23/2023] [Indexed: 07/24/2023]
Abstract
Transgenic models are useful tools for studying the pathogenesis of and drug development for Alzheimer's Disease (AD). AD models are constructed usually using overexpression or knock-in of multiple pathogenic gene mutations from familial AD. Each transgenic model has its unique behavioral and pathological features. This review summarizes the research progress of transgenic mouse models, and their progress in the unique mechanism of amyloid-β oligomers, including the first transgenic mouse model built in China based on a single gene mutation (PSEN1 V97L) found in Chinese familial AD. We further summarized the preclinical findings of drugs using the models, and their future application in exploring the upstream mechanisms and multitarget drug development in AD.
Collapse
Affiliation(s)
- Xinyue Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Wei Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- National Medical Center for Neurological Diseases and National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, 100053, China.
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
30
|
Hooshmandi E, Akbari S, Pandamooz S, Ghobadi M, Ghasemi R, Maghsoudi N, Rai SN, Borhani-Haghighi A, Salehi MS, Azarpira N, YousefiNejad A, Haghani M, Bayat M. Combined use of hair follicle stem cells and CEPO (carbamylated erythropoietin)-Fc in a rat model of chronic cerebral hypoperfusion: A behavioral, electrophysiological, and molecular study. Behav Brain Res 2023; 454:114655. [PMID: 37666305 DOI: 10.1016/j.bbr.2023.114655] [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: 07/02/2023] [Revised: 08/22/2023] [Accepted: 08/31/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND In dementia, synaptic dysfunction appears before neuronal loss. Stem cell therapy could potentially provide a promising strategy for the treatment of dementia models. The carbamylated erythropoietin fusion protein (CEPO-Fc) has shown synaptotrophic effects. This study aimed to determine the efficiency of the combined use of hair follicle stem cells (HFSC) and CEPO-Fc in the basal synaptic transmission (BST) and long-term plasticity (LTP) of chronic cerebral hypoperfusion (CCH) rats. METHODS We divided 64 adult rats into control, sham, CCH+vehicle, CCH+CEPO, CCH+HFSC, and CCH+HFSC+CEPO groups. The CEPO-Fc was injected three times/week for 30 days. HFSC transplantation was done on days 4, 14, and 21 after surgery. The Morris water maze test and passive avoidance were used to assess memory. BST and LTP were assessed by a field-potential recording of the CA1 region. The hippocampal mRNA expression of IGF-1, TGF-β1, β1-Catenine, NR2B, PSD-95, and GSk-3β was evaluated by quantitative RT-PCR. RESULTS Following combination therapy, spatial memory retention, and BST showed significant improvement relative to HFSC and CEPO-Fc groups. These effects were also confirmed by recovered mRNA expression of β1-catenin, TGF-β1, and NR2B. GSK-3β expression was downregulated in all treatment groups. The upregulated PSD-95 was identified in HFSC and combination groups compared to the vehicle group. CONCLUSIONS These findings indicate that the combined use of HFSC and CEPO-Fc may be more advantageous for treating memory disruption in the CCH model than CEPO-Fc or HFSC alone. This type of combination therapy may hopefully lead to a new approach to treatment for dementia.
Collapse
Affiliation(s)
- Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Somayeh Akbari
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran; Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Mojtaba Ghobadi
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Rasoul Ghasemi
- Neurophysiology Research Center and Physiology Department, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Nader Maghsoudi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | | | - Afshin Borhani-Haghighi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Mohammad Rasoul-Allah Research Tower, Shiraz, the Islamic Republic of Iran
| | - Amirhossein YousefiNejad
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Masoud Haghani
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran; Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran
| | - Mahnaz Bayat
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, the Islamic Republic of Iran.
| |
Collapse
|
31
|
Grieco SF, Holmes TC, Xu X. Probing neural circuit mechanisms in Alzheimer's disease using novel technologies. Mol Psychiatry 2023; 28:4407-4420. [PMID: 36959497 PMCID: PMC10827671 DOI: 10.1038/s41380-023-02018-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/25/2023]
Abstract
The study of Alzheimer's Disease (AD) has traditionally focused on neuropathological mechanisms that has guided therapies that attenuate neuropathological features. A new direction is emerging in AD research that focuses on the progressive loss of cognitive function due to disrupted neural circuit mechanisms. Evidence from humans and animal models of AD show that dysregulated circuits initiate a cascade of pathological events that culminate in functional loss of learning, memory, and other aspects of cognition. Recent progress in single-cell, spatial, and circuit omics informs this circuit-focused approach by determining the identities, locations, and circuitry of the specific cells affected by AD. Recently developed neuroscience tools allow for precise access to cell type-specific circuitry so that their functional roles in AD-related cognitive deficits and disease progression can be tested. An integrated systems-level understanding of AD-associated neural circuit mechanisms requires new multimodal and multi-scale interrogations that longitudinally measure and/or manipulate the ensemble properties of specific molecularly-defined neuron populations first susceptible to AD. These newly developed technological and conceptual advances present new opportunities for studying and treating circuits vulnerable in AD and represent the beginning of a new era for circuit-based AD research.
Collapse
Affiliation(s)
- Steven F Grieco
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
| | - Todd C Holmes
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping (CNCM), University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
32
|
Gastelum S, Michael AF, Bolger TA. Saccharomyces cerevisiae as a research tool for RNA-mediated human disease. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 15:e1814. [PMID: 37671427 DOI: 10.1002/wrna.1814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 09/07/2023]
Abstract
The budding yeast, Saccharomyces cerevisiae, has been used for decades as a powerful genetic tool to study a broad spectrum of biological topics. With its ease of use, economic utility, well-studied genome, and a highly conserved proteome across eukaryotes, it has become one of the most used model organisms. Due to these advantages, it has been used to study an array of complex human diseases. From broad, complex pathological conditions such as aging and neurodegenerative disease to newer uses such as SARS-CoV-2, yeast continues to offer new insights into how cellular processes are affected by disease and how affected pathways might be targeted in therapeutic settings. At the same time, the roles of RNA and RNA-based processes have become increasingly prominent in the pathology of many of these same human diseases, and yeast has been utilized to investigate these mechanisms, from aberrant RNA-binding proteins in amyotrophic lateral sclerosis to translation regulation in cancer. Here we review some of the important insights that yeast models have yielded into the molecular pathology of complex, RNA-based human diseases. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Stephanie Gastelum
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, USA
| | - Allison F Michael
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Timothy A Bolger
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| |
Collapse
|
33
|
Vallés AS, Barrantes FJ. Nicotinic Acetylcholine Receptor Dysfunction in Addiction and in Some Neurodegenerative and Neuropsychiatric Diseases. Cells 2023; 12:2051. [PMID: 37626860 PMCID: PMC10453526 DOI: 10.3390/cells12162051] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/20/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
The cholinergic system plays an essential role in brain development, physiology, and pathophysiology. Herein, we review how specific alterations in this system, through genetic mutations or abnormal receptor function, can lead to aberrant neural circuitry that triggers disease. The review focuses on the nicotinic acetylcholine receptor (nAChR) and its role in addiction and in neurodegenerative and neuropsychiatric diseases and epilepsy. Cholinergic dysfunction is associated with inflammatory processes mainly through the involvement of α7 nAChRs expressed in brain and in peripheral immune cells. Evidence suggests that these neuroinflammatory processes trigger and aggravate pathological states. We discuss the preclinical evidence demonstrating the therapeutic potential of nAChR ligands in Alzheimer disease, Parkinson disease, schizophrenia spectrum disorders, and in autosomal dominant sleep-related hypermotor epilepsy. PubMed and Google Scholar bibliographic databases were searched with the keywords indicated below.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Bahía Blanca Institute of Biochemical Research (UNS-CONICET), Bahía Blanca 8000, Argentina;
| | - Francisco J. Barrantes
- Biomedical Research Institute (BIOMED), Faculty of Medical Sciences, Pontifical Catholic University of Argentina—National Scientific and Technical Research Council, Av. Alicia Moreau de Justo 1600, Buenos Aires C1107AFF, Argentina
| |
Collapse
|
34
|
Fong V, Kanuri B, Traubert O, Lui M, Patel SB. Behavioral and metabolic and effects of ABCG4 KO in the APPswe,Ind (J9) mouse model of Alzheimer's disease. RESEARCH SQUARE 2023:rs.3.rs-3014093. [PMID: 37333297 PMCID: PMC10275060 DOI: 10.21203/rs.3.rs-3014093/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The pathogenesis of Alzheimer's disease (AD) is complex and involves an imbalance between production and clearance of amyloid-ß peptides (Aß), resulting in accumulation of Aß in senile plaques. Hypercholesterolemia is a major risk factor for developing AD, with cholesterol shown to accumulate in senile plaques and increase production of Aß. ABCG4 is a member of the ATP-binding cassette transporters predominantly expressed in the CNS, and has been suggested to play a role in cholesterol and Aß efflux from the brain. In this study, we bred Abcg4 knockout (KO) with the APPSwe,Ind (J9) mouse model of AD to test the hypothesis that loss of Abcg4 would exacerbate the AD phenotype. Unexpectedly, no differences were observed in Novel object recognition (NOR) and Novel object placement (NOP) behavioral tests, or on histologic examinations of brain tissues for senile plaque numbers. Furthermore, clearance of radiolabeled Aß from the brains did not differ between Abcg4 KO and control mice. Metabolic testing by indirect calorimetry, glucose tolerance test (GTT) and insulin tolerance test (ITT), were also mostly similar between groups with only a few mild metabolic differences noted. Overall these data suggest that the loss of ABCG4 did not exacerbate the AD phenotype.
Collapse
Affiliation(s)
- Vincent Fong
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati
| | - Babunageswararao Kanuri
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati
| | - Owen Traubert
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati
| | - Min Lui
- Department of Pathology & Laboratory Medicine, University of Cincinnati
| | - Shailendra B Patel
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Cincinnati
| |
Collapse
|
35
|
León R, Gutiérrez DA, Pinto C, Morales C, de la Fuente C, Riquelme C, Cortés BI, González-Martin A, Chamorro D, Espinosa N, Fuentealba P, Cancino GI, Zanlungo S, Dulcey AE, Marugan JJ, Álvarez Rojas A. c-Abl tyrosine kinase down-regulation as target for memory improvement in Alzheimer's disease. Front Aging Neurosci 2023; 15:1180987. [PMID: 37358955 PMCID: PMC10289333 DOI: 10.3389/fnagi.2023.1180987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/12/2023] [Indexed: 06/28/2023] Open
Abstract
Background Growing evidence suggests that the non-receptor tyrosine kinase, c-Abl, plays a significant role in the pathogenesis of Alzheimer's disease (AD). Here, we analyzed the effect of c-Abl on the cognitive performance decline of APPSwe/PSEN1ΔE9 (APP/PS1) mouse model for AD. Methods We used the conditional genetic ablation of c-Abl in the brain (c-Abl-KO) and pharmacological treatment with neurotinib, a novel allosteric c-Abl inhibitor with high brain penetrance, imbued in rodent's chow. Results We found that APP/PS1/c-Abl-KO mice and APP/PS1 neurotinib-fed mice had improved performance in hippocampus-dependent tasks. In the object location and Barnes-maze tests, they recognized the displaced object and learned the location of the escape hole faster than APP/PS1 mice. Also, APP/PS1 neurotinib-fed mice required fewer trials to reach the learning criterion in the memory flexibility test. Accordingly, c-Abl absence and inhibition caused fewer amyloid plaques, reduced astrogliosis, and preserved neurons in the hippocampus. Discussion Our results further validate c-Abl as a target for AD, and the neurotinib, a novel c-Abl inhibitor, as a suitable preclinical candidate for AD therapies.
Collapse
Affiliation(s)
- Rilda León
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela A. Gutiérrez
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudio Pinto
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian Morales
- Laboratory for Brain-Machine Interfaces and Neuromodulation, Facultad de Ingeniería, Instituto de Ingeniería Biológica y Médica, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory of Neural Circuits, Department of Psychiatry, Neuroscience Interdisciplinary Centre, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina de la Fuente
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristóbal Riquelme
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bastián I. Cortés
- Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Adrián González-Martin
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David Chamorro
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nelson Espinosa
- Laboratory of Neural Circuits, Department of Psychiatry, Neuroscience Interdisciplinary Centre, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Fuentealba
- Laboratory of Neural Circuits, Department of Psychiatry, Neuroscience Interdisciplinary Centre, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo I. Cancino
- Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Silvana Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Andrés E. Dulcey
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, United States
| | - Juan J. Marugan
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, United States
| | - Alejandra Álvarez Rojas
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
36
|
Van Skike CE, DeRosa N, Galvan V, Hussong SA. Rapamycin restores peripheral blood flow in aged mice and in mouse models of atherosclerosis and Alzheimer's disease. GeroScience 2023; 45:1987-1996. [PMID: 37052770 PMCID: PMC10400743 DOI: 10.1007/s11357-023-00786-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 03/19/2023] [Indexed: 04/14/2023] Open
Abstract
Peripheral artery disease (PAD), defined as reduced blood flow to the lower limbs, is a serious disorder that can lead to loss of function in the lower extremities and even loss of limbs. One of the main risk factors for PAD is age, with up to 25% of adults over the age of 55 and up to 40% over the age of 80 presenting with some form of the disease. While age is the largest risk factor for PAD, other risk factors include atherosclerosis, smoking, hypertension, and diabetes. Furthermore, previous studies have suggested that the incidence of PAD is significantly increased in patients with Alzheimer's disease (AD). Attenuation of mTOR with rapamycin significantly improves cerebral blood flow and heart function in aged rodents as well as in mouse models of atherosclerosis, atherosclerosis-driven cognitive impairment, and AD. In this study, we show that rapamycin treatment improves peripheral blood flow in aged mice and in mouse models of atherosclerosis and AD. Inhibition of mTOR with rapamycin ameliorates deficits in baseline hind paw perfusion in aged mice and restores levels of blood flow to levels indistinguishable from those of young controls. Furthermore, rapamycin treatment ameliorates peripheral blood flow deficits in mouse models of atherosclerosis and AD. These data indicate that mTOR is causally involved in the reduction of blood flow to lower limbs associated with aging, atherosclerosis, and AD-like progression in model mice. Rapamycin or other mTOR inhibitors may have potential as interventions to treat peripheral artery disease and other peripheral circulation-related conditions.
Collapse
Affiliation(s)
- Candice E Van Skike
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX, 78229, USA
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center San Antonio, San Antonio, TX, 78229, USA
| | - Nicholas DeRosa
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX, 78229, USA
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center San Antonio, San Antonio, TX, 78229, USA
| | - Veronica Galvan
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Health Care System, Oklahoma City, OK, 73104, USA.
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Stacy A Hussong
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Oklahoma City Veterans Health Care System, Oklahoma City, OK, 73104, USA
| |
Collapse
|
37
|
Bradberry MM, Peters-Clarke TM, Shishkova E, Chapman ER, Coon JJ. N-glycoproteomics of brain synapses and synaptic vesicles. Cell Rep 2023; 42:112368. [PMID: 37036808 PMCID: PMC10560701 DOI: 10.1016/j.celrep.2023.112368] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/13/2023] [Accepted: 03/23/2023] [Indexed: 04/11/2023] Open
Abstract
At mammalian neuronal synapses, synaptic vesicle (SV) glycoproteins are essential for robust neurotransmission. Asparagine (N)-linked glycosylation is required for delivery of the major SV glycoproteins synaptophysin and SV2A to SVs. Despite this key role for N-glycosylation, the molecular compositions of SV N-glycans are largely unknown. In this study, we combined organelle isolation techniques and high-resolution mass spectrometry to characterize N-glycosylation at synapses and SVs from mouse brain. Detecting over 2,500 unique glycopeptides, we found that SVs harbor a distinct population of oligomannose and highly fucosylated N-glycans. Using complementary fluorescence methods, we identify at least one highly fucosylated N-glycan enriched in SVs compared with synaptosomes. High fucosylation was characteristic of SV proteins, plasma membrane proteins, and cell adhesion molecules with key roles in synaptic function and development. Our results define the N-glycoproteome of a specialized neuronal organelle and inform timely questions in the glycobiology of synaptic pruning and neuroinflammation.
Collapse
Affiliation(s)
- Mazdak M Bradberry
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA; Howard Hughes Medical Institute and Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Trenton M Peters-Clarke
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA
| | - Evgenia Shishkova
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA
| | - Edwin R Chapman
- Howard Hughes Medical Institute and Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Joshua J Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; National Center for Quantitative Biology of Complex Systems, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53715, USA
| |
Collapse
|
38
|
Zott B, Konnerth A. Impairments of glutamatergic synaptic transmission in Alzheimer's disease. Semin Cell Dev Biol 2023; 139:24-34. [PMID: 35337739 DOI: 10.1016/j.semcdb.2022.03.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 12/31/2022]
Abstract
One of the hallmarks of Alzheimer's disease (AD) is structural cell damage and neuronal death in the brains of affected individuals. As these changes are irreversible, it is important to understand their origins and precursors in order to develop treatment strategies against AD. Here, we review evidence for AD-specific impairments of glutamatergic synaptic transmission by relating evidence from human AD subjects to functional studies in animal models of AD. The emerging picture is that early in the disease, the accumulation of toxic β-amyloid aggregates, particularly dimers and low molecular weight oligomers, disrupts glutamate reuptake, which leads to its extracellular accumulation causing neuronal depolarization. This drives the hyperactivation of neurons and might facilitate neuronal damage and degeneration through glutamate neurotoxicity.
Collapse
Affiliation(s)
- Benedikt Zott
- Institute of Neuroscience, Technical University of Munich, 80802 Munich, Germany; Munich Cluster for Systems Neurology, Technical University of Munich, 80802 Munich, Germany; Department of Neuroradiology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany.
| | - Arthur Konnerth
- Institute of Neuroscience, Technical University of Munich, 80802 Munich, Germany; Munich Cluster for Systems Neurology, Technical University of Munich, 80802 Munich, Germany
| |
Collapse
|
39
|
Nho K, Risacher SL, Apostolova L, Bice PJ, Brosch J, Deardorff R, Faber K, Farlow MR, Foroud T, Gao S, Rosewood T, Kim JP, Nudelman K, Yu M, Aisen P, Sperling R, Hooli B, Shcherbinin S, Svaldi D, Jack CR, Jagust WJ, Landau S, Vasanthakumar A, Waring JF, Doré V, Laws SM, Masters CL, Porter T, Rowe CC, Villemagne VL, Dumitrescu L, Hohman TJ, Libby JB, Mormino E, Buckley RF, Johnson K, Yang HS, Petersen RC, Ramanan VK, Vemuri P, Cohen AD, Fan KH, Kamboh MI, Lopez OL, Bennett DA, Ali M, Benzinger T, Cruchaga C, Hobbs D, De Jager PL, Fujita M, Jadhav V, Lamb BT, Tsai AP, Castanho I, Mill J, Weiner MW, Saykin AJ. Novel CYP1B1-RMDN2 Alzheimer's disease locus identified by genome-wide association analysis of cerebral tau deposition on PET. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.27.23286048. [PMID: 36993271 PMCID: PMC10055458 DOI: 10.1101/2023.02.27.23286048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Determining the genetic architecture of Alzheimer's disease (AD) pathologies can enhance mechanistic understanding and inform precision medicine strategies. Here, we performed a genome-wide association study of cortical tau quantified by positron emission tomography in 3,136 participants from 12 independent studies. The CYP1B1-RMDN2 locus was associated with tau deposition. The most significant signal was at rs2113389, which explained 4.3% of the variation in cortical tau, while APOE4 rs429358 accounted for 3.6%. rs2113389 was associated with higher tau and faster cognitive decline. Additive effects, but no interactions, were observed between rs2113389 and diagnosis, APOE4 , and Aβ positivity. CYP1B1 expression was upregulated in AD. rs2113389 was associated with higher CYP1B1 expression and methylation levels. Mouse model studies provided additional functional evidence for a relationship between CYP1B1 and tau deposition but not Aβ. These results may provide insight into the genetic basis of cerebral tau and novel pathways for therapeutic development in AD.
Collapse
|
40
|
Lin NH, Goh A, Lin SH, Chuang KA, Chang CH, Li MH, Lu CH, Chen WY, Wei PH, Pan IH, Perng MD, Wen SF. Neuroprotective Effects of a Multi-Herbal Extract on Axonal and Synaptic Disruption in Vitro and Cognitive Impairment in Vivo. J Alzheimers Dis Rep 2023; 7:51-76. [PMID: 36777330 PMCID: PMC9912829 DOI: 10.3233/adr-220056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
Background Alzheimer's disease (AD) is a multifactorial disorder characterized by cognitive decline. Current available therapeutics for AD have limited clinical benefit. Therefore, preventive therapies for interrupting the development of AD are critically needed. Molecules targeting multifunction to interact with various pathlogical components have been considered to improve the therapeutic efficiency of AD. In particular, herbal medicines with multiplicity of actions produce cognitive benefits on AD. Bugu-M is a multi-herbal extract composed of Ganoderma lucidum (Antler form), Nelumbo nucifera Gaertn., Ziziphus jujuba Mill., and Dimocarpus longan, with the ability of its various components to confer resilience to cognitive deficits. Objective To evaluate the potential of Bugu-M on amyloid-β (Aβ) toxicity and its in vitro mechanisms and on in vivo cognitive function. Methods We illustrated the effect of Bugu-M on Aβ25-35-evoked toxicity as well as its possible mechanisms to diminish the pathogenesis of AD in rat cortical neurons. For cognitive function studies, 2-month-old female 3×Tg-AD mice were administered 400 mg/kg Bugu-M for 30 days. Behavioral tests were performed to assess the efficacy of Bugu-M on cognitive impairment. Results In primary cortical neuronal cultures, Bugu-M mitigated Aβ-evoked toxicity by reducing cytoskeletal aberrations and axonal disruption, restoring presynaptic and postsynaptic protein expression, suppressing mitochondrial damage and apoptotic signaling, and reserving neurogenic and neurotrophic factors. Importantly, 30-day administration of Bugu-M effectively prevented development of cognitive impairment in 3-month-old female 3×Tg-AD mice. Conclusion Bugu-M might be beneficial in delaying the progression of AD, and thus warrants consideration for its preventive potential for AD.
Collapse
Affiliation(s)
- Ni-Hsuan Lin
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Angela Goh
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Shyh-Horng Lin
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Kai-An Chuang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chih-Hsuan Chang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Ming-Han Li
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chu-Hsun Lu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Wen-Yin Chen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Pei-Hsuan Wei
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - I-Hong Pan
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Ming-Der Perng
- Institute of Molecular Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan,
School of Medicine, College of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan,Correspondence to: Shu-Fang Wen, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, 321, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35743946; E-mail: and Ming-Der Perng, College of Life Sciences, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35742024; E-mail:
| | - Shu-Fang Wen
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan,Correspondence to: Shu-Fang Wen, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, 321, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35743946; E-mail: and Ming-Der Perng, College of Life Sciences, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu 300044, Taiwan. Tel.: +886 35742024; E-mail:
| |
Collapse
|
41
|
Das S, Datta S, Ghosal A, Chaudhuri NR, Sundaram G, Basu S. Screening of BACE1 inhibitors with antiamyloidogenic activity: A study of flavonoids and flavonoid derivatives. Neurosci Lett 2023; 792:136965. [PMID: 36379389 DOI: 10.1016/j.neulet.2022.136965] [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: 04/14/2022] [Revised: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Aggregates of β-amyloid peptide are found to occur in brains of AD patients and are formed upon sequential cleavage of the amyloid precursor protein by BACE1 and γ-secretase. Strategies inhibiting either peptide aggregation or the rate limiting enzyme BACE1 have been in demand for its implication in AD therapeutics. The present study is undertaken to mine compounds with dual ability. In this context, some natural compounds that were already predicted as BACE1 inhibitors by our group, were further tested for their activity as aggregation inhibitors. A pharmacophore model built with known antiamyloidogenic compounds was then applied for screening the natural compounds previously predicted as BACE1 inhibitors. Subsequently experimental validation by Thioflavin-T and Aβ-GFP assay filtered four compounds genistein, syringetin, tamarixetin and ZINC53276039. Out of them, ZINC53276039 showed promising antiamyloidogenic activity to act as a potent inhibitor of aggregation. Interestingly, our previous study revealed syringetin and ZINC53276039 to be good BACE1 inhibitors while tamarixetin to be a moderate BACE1 inhibitor. These good to moderate BACE1 inhibitors with moderate to reasonable antiamyloidogenic activity might show potency in reducing the amyloid load of AD brains.
Collapse
Affiliation(s)
- Sucharita Das
- Department of Microbiology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Suchismita Datta
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Agamani Ghosal
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Nibedita Ray Chaudhuri
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Geetanjali Sundaram
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| | - Soumalee Basu
- Department of Microbiology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| |
Collapse
|
42
|
Shea D, Colasurdo E, Smith A, Paschall C, Jayadev S, Keene CD, Galasko D, Ko A, Li G, Peskind E, Daggett V. SOBA: Development and testing of a soluble oligomer binding assay for detection of amyloidogenic toxic oligomers. Proc Natl Acad Sci U S A 2022; 119:e2213157119. [PMID: 36490316 PMCID: PMC9897489 DOI: 10.1073/pnas.2213157119] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/17/2022] [Indexed: 12/13/2022] Open
Abstract
The formation of toxic Amyloid β-peptide (Aβ) oligomers is one of the earliest events in the molecular pathology of Alzheimer's Disease (AD). These oligomers lead to a variety of downstream effects, including impaired neuronal signaling, neuroinflammation, tau phosphorylation, and neurodegeneration, and it is estimated that these events begin 10 to 20 y before the presentation of symptoms. Toxic Aβ oligomers contain a nonstandard protein structure, termed α-sheet, and designed α-sheet peptides target this main-chain structure in toxic oligomers independent of sequence. Here we show that a designed α-sheet peptide inhibits the deleterious effects on neuronal signaling and also serves as a capture agent in our soluble oligomer binding assay (SOBA). Pre-incubated synthetic α-sheet-containing Aβ oligomers produce strong SOBA signals, while monomeric and β-sheet protofibrillar Aβ do not. α-sheet containing oligomers were also present in cerebrospinal fluid (CSF) from an AD patient versus a noncognitively impaired control. For the detection of toxic oligomers in plasma, we developed a plate coating to increase the density of the capture peptide. The proof of concept was achieved by testing 379 banked human plasma samples. SOBA detected Aβ oligomers in patients on the AD continuum, including controls who later progressed to mild cognitive impairment. In addition, SOBA discriminated AD from other forms of dementia, yielding sensitivity and specificity of 99% relative to clinical and neuropathological diagnoses. To explore the broader potential of SOBA, we adapted the assay for a-synuclein oligomers and confirmed their presence in CSF from patients with Parkinson's disease and Lewy body dementia.
Collapse
Affiliation(s)
- Dylan Shea
- Molecular Engineering Program, University of Washington, Seattle, WA98105
- Department of Bioengineering, University of Washington, Seattle, WA98105
| | - Elizabeth Colasurdo
- Veterans Affairs Northwest Network Mental Illness Research, Education, and Clinical Center, Veteran Affairs Puget Sound Health Care System, Seattle, WA98108
| | - Alec Smith
- Department of Physiology and Biophysics, University of Washington, Seattle, 98105
| | - Courtnie Paschall
- Department of Bioengineering, University of Washington, Seattle, WA98105
- Department of Neurosurgery, University of Washington, Seattle, WA98195
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA98195
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA98195
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA92093
| | - Andrew Ko
- Department of Neurosurgery, University of Washington, Seattle, WA98195
| | - Ge Li
- Mental Illness Research, Education and Clinical Center Veterans Affairs Puget Sound Health Care System, Seattle, WA98108
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA98108
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA98195
| | - Elaine Peskind
- Veterans Affairs Northwest Network Mental Illness Research, Education, and Clinical Center, Veteran Affairs Puget Sound Health Care System, Seattle, WA98108
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA98195
| | - Valerie Daggett
- Molecular Engineering Program, University of Washington, Seattle, WA98105
- Department of Bioengineering, University of Washington, Seattle, WA98105
| |
Collapse
|
43
|
Chockanathan U, Padmanabhan K. From synapses to circuits and back: Bridging levels of understanding in animal models of Alzheimer's disease. Eur J Neurosci 2022; 56:5564-5586. [PMID: 35244297 PMCID: PMC10926359 DOI: 10.1111/ejn.15636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/04/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by behavioural changes that include memory loss and cognitive decline and is associated with the appearance of amyloid-β plaques and neurofibrillary tangles throughout the brain. Although aspects of the disease percolate across multiple levels of neuronal organization, from the cellular to the behavioural, it is increasingly clear that circuits are a critical junction between the cellular pathology and the behavioural phenotypes that bookend these levels of analyses. In this review, we discuss critical aspects of neural circuit research, beginning with synapses and progressing to network activity and how they influence our understanding of disease processed in AD.
Collapse
Affiliation(s)
- Udaysankar Chockanathan
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Neuroscience Graduate Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Krishnan Padmanabhan
- Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Neuroscience Graduate Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Ernest J. Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Center for Visual Science, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Intellectual and Developmental Disabilities Research Center, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
44
|
Mishra R, Phan T, Kumar P, Morrissey Z, Gupta M, Hollands C, Shetti A, Lopez KL, Maienschein-Cline M, Suh H, Hen R, Lazarov O. Augmenting neurogenesis rescues memory impairments in Alzheimer's disease by restoring the memory-storing neurons. J Exp Med 2022; 219:e20220391. [PMID: 35984475 PMCID: PMC9399756 DOI: 10.1084/jem.20220391] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/16/2022] [Accepted: 06/28/2022] [Indexed: 12/04/2022] Open
Abstract
Hippocampal neurogenesis is impaired in Alzheimer's disease (AD) patients and familial Alzheimer's disease (FAD) mouse models. However, it is unknown whether new neurons play a causative role in memory deficits. Here, we show that immature neurons were actively recruited into the engram following a hippocampus-dependent task. However, their recruitment is severely deficient in FAD. Recruited immature neurons exhibited compromised spine density and altered transcript profile. Targeted augmentation of neurogenesis in FAD mice restored the number of new neurons in the engram, the dendritic spine density, and the transcription signature of both immature and mature neurons, ultimately leading to the rescue of memory. Chemogenetic inactivation of immature neurons following enhanced neurogenesis in AD, reversed mouse performance, and diminished memory. Notably, AD-linked App, ApoE, and Adam10 were of the top differentially expressed genes in the engram. Collectively, these observations suggest that defective neurogenesis contributes to memory failure in AD.
Collapse
Affiliation(s)
- Rachana Mishra
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Trongha Phan
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Pavan Kumar
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Zachery Morrissey
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
- Department of Psychiatry, College of Medicine, The University of Illinois at Chicago, Chicago, IL
- The Graduate Program in Neuroscience, The University of Illinois at Chicago, Chicago, IL
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Carolyn Hollands
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Aashutosh Shetti
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | - Kyra Lauren Lopez
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| | | | - Hoonkyo Suh
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH
| | - Rene Hen
- Department of Psychiatry, Irving Medical Center, Columbia University, New York, NY
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
45
|
Yang H, Li J, Li X, Ma L, Hou M, Zhou H, Zhou R. Based on molecular structures: Amyloid-β generation, clearance, toxicity and therapeutic strategies. Front Mol Neurosci 2022; 15:927530. [PMID: 36117918 PMCID: PMC9470852 DOI: 10.3389/fnmol.2022.927530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Amyloid-β (Aβ) has long been considered as one of the most important pathogenic factors in Alzheimer’s disease (AD), but the specific pathogenic mechanism of Aβ is still not completely understood. In recent years, the development of structural biology technology has led to new understandings about Aβ molecular structures, Aβ generation and clearance from the brain and peripheral tissues, and its pathological toxicity. The purpose of the review is to discuss Aβ metabolism and toxicity, and the therapeutic strategy of AD based on the latest progress in molecular structures of Aβ. The Aβ structure at the atomic level has been analyzed, which provides a new and refined perspective to comprehend the role of Aβ in AD and to formulate therapeutic strategies of AD.
Collapse
Affiliation(s)
- Hai Yang
- Department of Neurology, Army Medical Center of PLA, Chongqing, China
| | - Jinping Li
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xiaoxiong Li
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Linqiu Ma
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Mingliang Hou
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Huadong Zhou
- Department of Neurology, Army Medical Center of PLA, Chongqing, China
| | - Rui Zhou
- Southwest Hospital, Army Medical University, Chongqing, China
- *Correspondence: Rui Zhou,
| |
Collapse
|
46
|
Diaz A, Martin-Jimenez C, Woo Y, Merino P, Torre E, Yepes M. Urokinase-Type Plasminogen Activator Triggers Wingless/Int1-Independent Phosphorylation of the Low-Density Lipoprotein Receptor-Related Protein-6 in Cerebral Cortical Neurons. J Alzheimers Dis 2022; 89:877-891. [PMID: 35964187 DOI: 10.3233/jad-220320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Urokinase-type plasminogen activator (uPA) is a serine proteinase found in excitatory synapses located in the II/III and V cortical layers. The synaptic release of uPA promotes the formation of synaptic contacts and the repair of synapses damaged by various forms of injury, and its abundance is decreased in the synapse of Alzheimer's disease (AD) patients. Inactivation of the Wingless/Int1 (Wnt)-β-catenin pathway plays a central role in the pathogenesis of AD. Soluble amyloid-β (Aβ) prevents the phosphorylation of the low-density lipoprotein receptor-related protein-6 (LRP6), and the resultant inactivation of the Wnt-β-catenin pathway prompts the amyloidogenic processing of the amyloid-β protein precursor (AβPP) and causes synaptic loss. OBJECTIVE To study the role of neuronal uPA in the pathogenesis of AD. METHODS We used in vitro cultures of murine cerebral cortical neurons, a murine neuroblastoma cell line transfected with the APP-695 Swedish mutation (N2asw), and mice deficient on either plasminogen, or uPA, or its receptor (uPAR). RESULTS We show that uPA activates the Wnt-β-catenin pathway in cerebral cortical neurons by triggering the phosphorylation of LRP6 via a plasmin-independent mechanism that does not require binding of Wnt ligands (Wnts). Our data indicate that uPA-induced activation of the Wnt-β-catenin pathway protects the synapse from the harmful effects of soluble Aβ and prevents the amyloidogenic processing of AβPP by inhibiting the expression of β-secretase 1 (BACE1) and the ensuing generation of Aβ 40 and Aβ 42 peptides. CONCLUSION uPA protects the synapse and antagonizes the inhibitory effect of soluble Aβ on the Wnt-β-catenin pathway by providing an alternative pathway for LRP6 phosphorylation and β-catenin stabilization.
Collapse
Affiliation(s)
- Ariel Diaz
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Cynthia Martin-Jimenez
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Yena Woo
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Paola Merino
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Enrique Torre
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Manuel Yepes
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA.,Department of Neurology, Emory University, Atlanta, GA, USA.,Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA
| |
Collapse
|
47
|
He A, Zhang C, Ke X, Yi Y, Yu Q, Zhang T, Yu H, Du H, Li H, Tian Q, Zhu LQ, Lu Y. VGLUT3 neurons in median raphe control the efficacy of spatial memory retrieval via ETV4 regulation of VGLUT3 transcription. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1590-1607. [PMID: 35089530 DOI: 10.1007/s11427-021-2047-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/15/2021] [Indexed: 06/14/2023]
Abstract
The raphe nucleus is critical for feeding, rewarding and memory. However, how the heterogenous raphe neurons are molecularly and structurally organized to engage their divergent functions remains unknown. Here, we genetically target a subset of neurons expressing VGLUT3. VGLUT3 neurons control the efficacy of spatial memory retrieval by synapsing directly with parvalbumin-expressing GABA interneurons (PGIs) in the dentate gyrus. In a mouse model of Alzheimer's disease (AD mice), VGLUT3→PGIs synaptic transmission is impaired by ETV4 inhibition of VGLUT3 transcription. ETV4 binds to a promoter region of VGLUT3 and activates VGLUT3 transcription in VGLUT3 neurons. Strengthening VGLUT3→PGIs synaptic transmission by ETV4 activation of VGLUT3 transcription upscales the efficacy of spatial memory retrieval in AD mice. This study reports a novel circuit and molecular mechanism underlying the efficacy of spatial memory retrieval via ETV4 inhibition of VGLUT3 transcription and hence provides a promising target for therapeutic intervention of the disease progression.
Collapse
Affiliation(s)
- Aodi He
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chen Zhang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Ke
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yao Yi
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Quntao Yu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tongmei Zhang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongyan Yu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huiyun Du
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hao Li
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qing Tian
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling-Qiang Zhu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Youming Lu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
48
|
Chocron ES, Munkácsy E, Kim HS, Karpowicz P, Jiang N, Van Skike CE, DeRosa N, Banh AQ, Palavicini JP, Wityk P, Kalinowski L, Galvan V, Osmulski PA, Jankowska E, Gaczynska M, Pickering AM. Genetic and pharmacologic proteasome augmentation ameliorates Alzheimer's-like pathology in mouse and fly APP overexpression models. SCIENCE ADVANCES 2022; 8:eabk2252. [PMID: 35675410 PMCID: PMC9177073 DOI: 10.1126/sciadv.abk2252] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 04/21/2022] [Indexed: 05/27/2023]
Abstract
The proteasome has key roles in neuronal proteostasis, including the removal of misfolded and oxidized proteins, presynaptic protein turnover, and synaptic efficacy and plasticity. Proteasome dysfunction is a prominent feature of Alzheimer's disease (AD). We show that prevention of proteasome dysfunction by genetic manipulation delays mortality, cell death, and cognitive deficits in fly and cell culture AD models. We developed a transgenic mouse with neuronal-specific proteasome overexpression that, when crossed with an AD mouse model, showed reduced mortality and cognitive deficits. To establish translational relevance, we developed a set of TAT-based proteasome-activating peptidomimetics that stably penetrated the blood-brain barrier and enhanced 20S/26S proteasome activity. These agonists protected against cell death, cognitive decline, and mortality in cell culture, fly, and mouse AD models. The protective effects of proteasome overexpression appear to be driven, at least in part, by the proteasome's increased turnover of the amyloid precursor protein along with the prevention of overall proteostatic dysfunction.
Collapse
Affiliation(s)
- E. Sandra Chocron
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
| | - Erin Munkácsy
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
| | - Harper S. Kim
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
- Center for Neurodegeneration and Experimental Therapeutics (CNET), Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Przemyslaw Karpowicz
- Department of Organic Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Nisi Jiang
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Candice E. Van Skike
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, USA
| | - Nicholas DeRosa
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, USA
| | - Andy Q. Banh
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Cellular and Integrative Physiology, UT Health San Antonio, San Antonio, TX, USA
| | - Juan P. Palavicini
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
| | - Paweł Wityk
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
- Department of Medical Laboratory Diagnostics–Fahrenheit Biobank BBMRI.pl, Medical University of Gdańsk, Gdańsk, Poland
- BioTechMed Centre/Department of Mechanics of Materials and Structures, Gdańsk University of Technology, Gdańsk, Poland
| | - Leszek Kalinowski
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Gdańsk, Poland
- Department of Medical Laboratory Diagnostics–Fahrenheit Biobank BBMRI.pl, Medical University of Gdańsk, Gdańsk, Poland
- BioTechMed Centre/Department of Mechanics of Materials and Structures, Gdańsk University of Technology, Gdańsk, Poland
| | - Veronica Galvan
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- College of Medicine, Oklahoma Health Science Center, Oklahoma City, OK, USA
- Department of Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- South Texas VA Health Care System, San Antonio, TX, USA
- Oklahoma City VA Health Care System, Oklahoma City, OK, USA
| | - Pawel A. Osmulski
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Elzbieta Jankowska
- Department of Organic Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Maria Gaczynska
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Andrew M. Pickering
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX, USA
- Center for Neurodegeneration and Experimental Therapeutics (CNET), Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
49
|
Theta and gamma oscillatory dynamics in mouse models of Alzheimer's disease: A path to prospective therapeutic intervention. Neurosci Biobehav Rev 2022; 136:104628. [PMID: 35331816 DOI: 10.1016/j.neubiorev.2022.104628] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 12/26/2022]
Abstract
Understanding the neural basis of cognitive deficits, a key feature of Alzheimer's disease (AD), is imperative for achieving the therapy of the disease. Rhythmic oscillatory activities in neural systems are a fundamental mechanism for diverse brain functions, including cognition. In several neurological conditions like AD, aberrant neural oscillations have been shown to play a central role. Furthermore, manipulation of brain oscillations in animals has confirmed their impact on cognition and disease. In this article, we review the evidence from mouse models that shows how synchronized oscillatory activity is intricately linked to AD machinery. We primarily focus on recent reports showing abnormal oscillatory activities at theta and gamma frequencies in AD condition and their influence on cellular disturbances and cognitive impairments. A thorough comprehension of the role that neuronal oscillations play in AD pathology should pave the way to therapeutic interventions that can curb the disease.
Collapse
|
50
|
Abstract
Alzheimer’s Disease (AD) is a neurodegenerative disorder that is characterized clinically by progressive cognitive decline and pathologically by the β-sheet rich fibril plaque deposition of the amyloid-β (Aβ) peptide in the brain. While plaques are a hallmark of AD, plaque burden is not correlated with cognitive impairment. Instead, Aβ oligomers formed during the aggregation process represent the main agents of neurotoxicity, which occurs 10–20 years before patients begin to show symptoms. These oligomers are dynamic in nature and represented by a heterogeneous distribution of aggregates ranging from low- to high-molecular weight, some of which are toxic while others are not. A major difficulty in determining the pathological mechanism(s) of Aβ, developing reliable diagnostic markers for early-stage detection, as well as effective therapeutics for AD are the differentiation and characterization of oligomers formed throughout disease propagation based on their molecular features, effects on biological function, and relevance to disease propagation and pathology. Thus, it is critical to methodically identify the mechanisms of Aβ aggregation and toxicity, as well as describe the roles of different oligomers and aggregates in disease progression and molecular pathology. Here, we describe a variety of biophysical techniques used to isolate and characterize a range of Aβ oligomer populations, as well as discuss proposed mechanisms of toxicity and therapeutic interventions aimed at specific assemblies formed during the aggregation process. The approaches being used to map the misfolding and aggregation of Aβ are like what was done during the fundamental early studies, mapping protein folding pathways using combinations of biophysical techniques in concert with protein engineering. Such information is critical to the design and molecular engineering of future diagnostics and therapeutics for AD.
Collapse
|