1
|
da Silva EA, Faber J, Penitente AR, Fernandes J, Bertolucci PHF, Longo BM, Arida RM. Effects of resistance exercise on behavioral and molecular changes in transgenic female mice for Alzheimer's disease in early and advanced stages. Exp Neurol 2025; 388:115217. [PMID: 40089002 DOI: 10.1016/j.expneurol.2025.115217] [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: 06/27/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that affects memory and cognition, with a higher prevalence in women. Given the lack of effective treatment, physical activity stands out as a complementary approach to prevent or delay disease progression. While numerous studies on humans and animals indicate that aerobic exercise induces brain changes, the impact of resistance exercise (RE) on AD is not fully understood. OBJECTIVE This study aimed to investigate the behavioral and molecular changes induced by RE in female transgenic mice with AD at the early and advanced stages of the disease. MATERIALS AND METHODS Adult (initial phase - 7 to 8 months of age, n = 32) and adult/elderly (advanced phase - 22 to 23 months of age, n = 32) female mice (2xTg-AD) for the APPSWE/PS1dE9 mutation were subjected to a four-week RE protocol. Mobility, anxiety-like behavior, long-term memory (LTM), and depressive-like behavior were assessed. Beta-amyloid (βA) and cytokines were quantified using the ELISA technique. RESULTS There was a progressive increase in strength in both trained groups at different ages. RE reversed memory deficits only in adult AD animals and the anxiety-like behavior only in adult/elderly AD animals. RE reversed depressive-like behavior in adult and adult/elderly AD animals. RE reduced βA only in adult AD animals. RE modified the expression of several cytokines in animals in the early and advanced stage of AD. CONCLUSION RE can be a promising strategy to minimize the deleterious effects of AD; however, its effectiveness may be more limited to the early stages of the disease.
Collapse
Affiliation(s)
| | - Jean Faber
- Federal University of Sao Paulo - Neurology and Neurosurgery Department, Brazil
| | | | | | | | | | | |
Collapse
|
2
|
Fatima G, Ashiquzzaman A, Kim SS, Kim YR, Kwon HS, Chung E. Vascular and glymphatic dysfunction as drivers of cognitive impairment in Alzheimer's disease: Insights from computational approaches. Neurobiol Dis 2025; 208:106877. [PMID: 40107629 DOI: 10.1016/j.nbd.2025.106877] [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: 01/16/2025] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025] Open
Abstract
Alzheimer's disease (AD) is driven by complex interactions between vascular dysfunction, glymphatic system impairment, and neuroinflammation. Vascular aging, characterized by arterial stiffness and reduced cerebral blood flow (CBF), disrupts the pulsatile forces necessary for glymphatic clearance, exacerbating amyloid-beta (Aβ) accumulation and cognitive decline. This review synthesizes insights into the mechanistic crosstalk between these systems and explores their contributions to AD pathogenesis. Emerging machine learning (ML) tools, such as DeepLabCut and Motion sequencing (MoSeq), offer innovative solutions for analyzing multimodal data and enhancing diagnostic precision. Integrating ML with imaging and behavioral analyses bridges gaps in understanding vascular-glymphatic dysfunction. Future research must prioritize these interactions to develop early diagnostics and targeted interventions, advancing our understanding of neurovascular health in AD.
Collapse
Affiliation(s)
- Gehan Fatima
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea
| | - Akm Ashiquzzaman
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea
| | - Sang Seong Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea
| | - Young Ro Kim
- Department of Radiology, Harvard Medical School, Boston, MA 02115, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Hyuk-Sang Kwon
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea; AI Graduate School, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Rep. of Korea; Research Center for Photon Science Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea.
| | - Euiheon Chung
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea; AI Graduate School, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Rep. of Korea; Research Center for Photon Science Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Rep. of Korea.
| |
Collapse
|
3
|
Wang LY, Hu H, Sheng ZH, Hu HY, Ou YN, Guo F, Zhu YK, Tan L. Associations among Angiotensin-Converting Enzyme, Neuroinflammation, and Cerebrospinal Fluid Biomarkers of Alzheimer's Disease in Non-Dementia Adults. Neurotox Res 2025; 43:20. [PMID: 40186068 DOI: 10.1007/s12640-025-00740-8] [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: 09/26/2024] [Revised: 02/12/2025] [Accepted: 03/16/2025] [Indexed: 04/07/2025]
Abstract
Recent studies have identified the angiotensin-converting enzyme (ACE) gene as a potential candidate influencing Alzheimer's disease (AD) risk. It is crucial to investigate the impact of ACE on AD pathology and its underlying mechanisms. A total of 450 non-demented participants from the Alzheimer's disease Neuroimaging Initiative (ADNI) with data on cerebrospinal fluid (CSF) ACE, AD core biomarkers and inflammation-related biomarkers were included. Multiple linear regression was used to assess the associations among CSF ACE, AD core biomarkers and inflammation-related biomarkers. And we used the mediation models to investigate the potential mechanisms through which ACE influenced AD pathology. The results of multiple linear regression were shown that CSF ACE was significantly correlated with CSF Aβ42, P-tau, T-tau (all P < 0.001), and inflammation-related biomarkers (soluble triggering receptor expressed on myeloid cells 2 [sTREM2], progranulin [PGRN], glial fibrillary acidic protein [GFAP], transforming growth factor [TGF]-β1, TGF-β2, TGF-β3, tumor necrosis factor [TNF]-R1, TNF-R2, TNF-α, interleukin [IL]-21, IL-6, IL-7, IL-9, IL-10, IL-12p40, vascular cell adhesion molecule-1 [VCAM-1], and intercellular adhesion molecule-1 [ICAM-1]) (all P < 0.05). In addition, the mediation analysis results showed that the association of CSF ACE and inflammation-related biomarkers (sTREM2, PGRN, TGF-β1, TGF-β2, TNFR1, IL-6, IL-7, IL-9, and VCAM-1) mediated the correlation of CSF Aβ42 with P-tau. Our findings show that CSF ACE and neuroinflammation are correlated and that their correlation mediates the link between Aβ pathology and P-tau. This suggests ACE may play a significant role in the progression from Aβ pathology to tau pathology.
Collapse
Affiliation(s)
- Lan-Yang Wang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Ze-Hu Sheng
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Fan Guo
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yang-Ke Zhu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Rullmann M, Henssen D, Melasch JT, Scherlach C, Saur D, Schroeter ML, Tiepolt S, Koglin N, Stephens AW, Hesse S, Strauss M, Brendel M, Mishchenko O, Schildan A, Classen J, Hoffmann KT, Sabri O, Barthel H. Multi-parametric [ 18F]PI-2620 tau PET/MRI for the phenotyping of different Alzheimer's disease variants. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07135-z. [PMID: 39937274 DOI: 10.1007/s00259-025-07135-z] [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: 01/10/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE Heterogeneity in clinical phenotypes has led to the description of different phenotypes of Alzheimer's disease (AD). Besides the most frequent amnestic variant of AD (aAD), patients presenting with language deficits are diagnosed with logopenic variant primary progressive aphasia (lvPPA), whereas patients presenting with visual deficits are classified as posterior cortical atrophy (PCA). METHODS This study set out to investigate the value of a multi-parametric [18F]PI-2620 tau PET/MRI protocol to distinguish aAD, lvPPA and PCA to support clinical diagnosis in 32 patients. Phenotype-specific information about tau accumulation, relative perfusion, grey matter density, functional network alterations and white matter microstructural alterations was collected. RESULTS The aAD patients showed significantly higher tau accumulation, relative hypoperfusion and grey matter density loss in the temporal lobes compared to PCA and lvPPA patients. PCA patients, on the other hand, showed significantly higher tau accumulation in the occipital lobe as compared to aAD patients. Relative hypoperfusion in the occipital lobe and loss of functional connectivity of the posterior cingulate cortex to supplementary visual cortical regions helped to distinguish PCA from lvPPA. Tau accumulation in the cerebellum and microstructural changes in the cingulum were found to help differentiate lvPPA from aAD. CONCLUSION This study highlights structural and functional differences between patients with different AD phenotypes. Differences in regional tau PET signals suggest that refinements in the Braak staging system are needed for the non-aAD cases. These patterns of tau accumulation align with the cascading network failure hypothesis, though more research is needed to warrant the here presented results in larger patient cohorts.
Collapse
Affiliation(s)
- Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany.
| | - Dylan Henssen
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Juliana T Melasch
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Cordula Scherlach
- Department of Neuroradiology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Dorothee Saur
- Department of Neurology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Matthias L Schroeter
- Clinic for Cognitive Neurology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | | | | | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Maria Strauss
- Department of Psychiatry, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Olena Mishchenko
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Andreas Schildan
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Joseph Classen
- Department of Neurology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Karl-Titus Hoffmann
- Department of Neuroradiology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
- Department of Nuclear Medicine, Hospital Dessau, Dessau, Germany
| |
Collapse
|
5
|
Pardo K, Khasminsky V, Keret O, Benninger F, Goldberg I, Shelef I, Auriel E, Glik A. Alzheimer's disease patients have smaller venous drainage system compared to cognitively healthy controls. Alzheimers Dement 2025; 21:e14551. [PMID: 39936326 PMCID: PMC11851167 DOI: 10.1002/alz.14551] [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: 10/09/2024] [Revised: 12/21/2024] [Accepted: 01/25/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION One of the pathological hallmarks of Alzheimer's disease (AD) is the accumulation of amyloid beta 42 (Aβ42). Decreased venous drainage may enhance Aβ42 accumulation. We aimed to compare venous cross-sectional area (CSA) of AD patients to cognitively healthy controls. METHODS All patients underwent neurocognitive evaluation and brain magnetic resonance imaging, including time-of-flight sequence. Venous CSA was measured at the jugular foramen level. RESULTS Thirty-nine AD/mild cognitive impairment patients and 20 cognitively healthy controls were included. Total venous CSA was smaller in the cognitively impaired group (mean CSA 139.77 mm2 [SD: 32.22] vs 166.55 mm2 [SD: 33.1], p = 0.004]. When divided, both internal jugular and non-jugular systems were smaller within cognitively impaired patients; statistical significance was achieved only for the non-jugular system (mean CSA 41.21 mm2 [SD: 21.52] vs 54.5 mm2 [SD: 27.31], p = 0.045). DISCUSSION There is an association between smaller venous systems and cognitive impairment, most prominently in the non-jugular system. Venous narrowing may cause impaired venous drainage, leading to an accumulation of Aβ42. HIGHLIGHTS The non-jugular venous system, including the vertebral plexus and pterygopalatine plexus, plays an important role in cerebral drainage. The total venous CSA is significantly smaller in cognitively impaired patients compared to healthy controls. Reduced venous drainage may contribute to the accumulation of Aβ and other waste products and potentially plays a role in AD pathology.
Collapse
Affiliation(s)
- Keshet Pardo
- Department of NeurologyRabin Medical Center – Beilinson HospitalPetach TikvaIsrael
- School of MedicineTel‐Aviv UniversityTel‐AvivIsrael
| | - Vadim Khasminsky
- School of MedicineTel‐Aviv UniversityTel‐AvivIsrael
- Department of RadiologyRabin Medical Center – Beilinson HospitalPetach TikvaIsrael
| | - Ophir Keret
- School of MedicineTel‐Aviv UniversityTel‐AvivIsrael
- Cognitive Neurology ServiceRabin Medical Center – Beilinson HospitalPetach TikvaIsrael
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Felix Benninger
- Department of NeurologyRabin Medical Center – Beilinson HospitalPetach TikvaIsrael
- School of MedicineTel‐Aviv UniversityTel‐AvivIsrael
| | - Ilan Goldberg
- Department of NeurologyRabin Medical Center – Beilinson HospitalPetach TikvaIsrael
- School of MedicineTel‐Aviv UniversityTel‐AvivIsrael
| | - Ilan Shelef
- Department of RadiologySoroka Medical Center Beer ShevaBeershebaIsrael
| | - Eitan Auriel
- Department of NeurologyRabin Medical Center – Beilinson HospitalPetach TikvaIsrael
- School of MedicineTel‐Aviv UniversityTel‐AvivIsrael
| | - Amir Glik
- School of MedicineTel‐Aviv UniversityTel‐AvivIsrael
- Cognitive Neurology ServiceRabin Medical Center – Beilinson HospitalPetach TikvaIsrael
| |
Collapse
|
6
|
Hossain MK, Chae HJ. Calcium balance through mutual orchestrated inter-organelle communication: A pleiotropic target for combating Alzheimer's disease. Neurochem Int 2025; 182:105905. [PMID: 39566580 DOI: 10.1016/j.neuint.2024.105905] [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: 07/06/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
Dysfunctional intraneuronal organelles in Alzheimer's Disease (AD) propel aberrant calcium handling, triggering molecular miscommunication within organelles such as mitochondria, endoplasmic reticulum, and lysosomes. This disruption in organelle function not only impairs cellular homeostasis but also exacerbates neurodegenerative processes involving the accumulation of amyloid-β (Aβ) and hyperphosphorylated tau, amplifying the disease's vicious cycle. In this review, the concept of Mutual Orchestrated Inter-organelle Communication (MOIC) proposes potential therapeutic avenues for restoring Ca2+ homeostasis in AD, offering a theoretical framework for developing disease-modifying treatments. The intricate nature of AD necessitates a shift towards combination therapies targeting MOIC-associated pathways, presenting a more effective approach than monotherapy.
Collapse
Affiliation(s)
| | - Han Jung Chae
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Republic of Korea.
| |
Collapse
|
7
|
Thai QM, Nguyen TH, Lenon GB, Thu Phung HT, Horng JT, Tran PT, Ngo ST. Estimating AChE inhibitors from MCE database by machine learning and atomistic calculations. J Mol Graph Model 2025; 134:108906. [PMID: 39561662 DOI: 10.1016/j.jmgm.2024.108906] [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: 02/02/2024] [Revised: 08/17/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024]
Abstract
Acetylcholinesterase (AChE) is one of the most successful targets for the treatment of Alzheimer's disease (AD). Inhibition of AChE can result in preventing AD. In this context, the machine-learning (ML) model, molecular docking, and molecular dynamics calculations were employed to characterize the potential inhibitors for AChE from MedChemExpress (MCE) database. The trained ML model was initially employed for estimating the inhibitory of MCE compounds. Atomistic simulations including molecular docking and molecular dynamics simulations were then used to confirm ML outcomes. In particular, the physical insights into the ligand binding to AChE were clarified over the calculations. Two compounds, PubChem ID of 130467298 and 132020434, were indicated that they can inhibit AChE.
Collapse
Affiliation(s)
- Quynh Mai Thai
- Laboratory of Biophysics, Institute for Advanced Study in Technology, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam.
| | - Trung Hai Nguyen
- Laboratory of Biophysics, Institute for Advanced Study in Technology, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam
| | - George Binh Lenon
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Huong Thi Thu Phung
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Viet Nam
| | - Jim-Tong Horng
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Phuong-Thao Tran
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, 008404, Viet Nam
| | - Son Tung Ngo
- Laboratory of Biophysics, Institute for Advanced Study in Technology, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam.
| |
Collapse
|
8
|
Asfour AAR, Evren AE, Sağlık Özkan BN, Yurttaş L. Investigating the potential of novel thiazole derivatives in treating Alzheimer's and Parkinson's diseases. J Biomol Struct Dyn 2024:1-17. [PMID: 39672098 DOI: 10.1080/07391102.2024.2437521] [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: 01/22/2024] [Accepted: 05/17/2024] [Indexed: 12/15/2024]
Abstract
The study aimed to investigate 12 novel thiazole compounds in the treatment of neurodegenerative disorders. The compounds produced were evaluated for their inhibitory efficacy against acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and monoamine oxidases (MAOs). Among the compounds, 5d, 5e, and 5j showed the highest AChE inhibitory activity. The IC50 values for compounds are 0.223 ± 0.010 µM, 0.092 ± 0.003 µM, and 0.054 ± 0.002 µM, respectively. In addition, molecular docking analyses and molecular dynamic simulation were used to examine the interactions of these compounds with protein sites. The results suggest that thiazole-ring compounds could serve as a promising basis for the development of drugs aimed at treating neurodegenerative diseases (NDD), caused by Parkinson's and Alzheimer's diseases.
Collapse
Affiliation(s)
- Abd Al Rahman Asfour
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Anadolu University, Eskişehir, Turkey
- Institute of Graduate Education, Department of Pharmaceutical Chemistry, Anadolu University, Eskişehir, Turkey
| | - Asaf Evrim Evren
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Anadolu University, Eskişehir, Turkey
- Department of Pharmacy Services, Bilecik Seyh Edebali University, Vocational School of Health Services, Bilecik
| | | | - Leyla Yurttaş
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
9
|
Thai QM, Tung NT, Do Thi Mai D, Ngo ST. Dimerization of the Aβ 42 under the Influence of the Gold Nanoparticle: A REMD Study. J Phys Chem B 2024; 128:11705-11713. [PMID: 39508442 DOI: 10.1021/acs.jpcb.4c06224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Advances in Alzheimer's disease (AD) are related to the oligomerization of Amyloid β (Aβ) peptides. Therefore, alteration of the process can prevent AD. We investigated the Aβ42 dimerization under the effects of gold nanoparticles using temperature replica-exchange molecular dynamics (REMD) simulations. The structural change of dimers in the presence and absence of the gold nanoparticle, Au55, was monitored over stable intervals. Physical insights into the oligomerization of Aβ were thus clarified. The computed metrics indicate that Au55 affects the progress of oligomerization. Specifically, the presence of the gold nanoparticle significantly modifies the structure of dimeric Aβ42. The β-content experienced a substantial decrease with the induction of Au55. The turn and coil-contents are also decreased under the effects of the gold nanoparticle. However, the α-content of the dimer exhibited a rigid increase. The influence of gold nanoparticles on the dimeric Aβ42 differs significantly from that of silver nanoparticles, which reduce β-content but increase coil-, turn-, and α-contents. The nature of inhibition will be discussed, in which the vdW interaction plays a driving force for the interaction between the Aβ42 dimer and the gold nanoparticle.
Collapse
Affiliation(s)
- Quynh Mai Thai
- Laboratory of Biophysics, Institute for Advanced Study in Technology, Ton Duc Thang University, Ho Chi Minh City 72915, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 72915, Vietnam
| | - Nguyen Thanh Tung
- Institute of Materials Science, Vietnam Academy of Science and Technology, Hanoi 11307, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi 11307, Vietnam
| | - Dung Do Thi Mai
- Faculty of Pharmaceutical Chemistry and Technology, Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 11021, Vietnam
| | - Son Tung Ngo
- Laboratory of Biophysics, Institute for Advanced Study in Technology, Ton Duc Thang University, Ho Chi Minh City 72915, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 72915, Vietnam
| |
Collapse
|
10
|
Arumugam D, Jamuna NA, Kamalakshan A, Mandal S. Modulation of AIE and Intramolecular Charge Transfer of a Pyrene-Based Probe for Discriminatory Detection and Imaging of Oligomers and Amyloid Fibrils. ACS APPLIED BIO MATERIALS 2024; 7:6343-6356. [PMID: 39291866 DOI: 10.1021/acsabm.4c00820] [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: 09/19/2024]
Abstract
Oligomers and amyloid fibrils formed at different stages of protein aggregation are important biomarkers for a variety of neurodegenerative diseases including Alzheimer's and Parkinson's diseases. The development of probes for the sensitive detection of oligomeric species is important for early stage diagnosis of amyloidogenic diseases. Many small molecular dyes have been developed to probe the dynamic growth of amyloid fibrils. However, there is a lack of discriminatory detection strategies to monitor the dynamics of both oligomers and amyloid fibrils based on the differential modulation of the photophysical properties of a single dye. Here we report a pyrene-based intramolecular charge transfer (ICT) dye with large Stokes shifted red-emitting aggregation induced emission (AIE) for monitoring the dynamic populations of both oligomers and fibrils during the aggregation of hen egg white lysozyme (HEWL) protein. At the early stage of protein aggregation, the accumulation of HEWL oligomers results in a rapid and substantial increase in the red AIE intensity at 660 nm. Later, as the oligomers transform into mature fibrils, the dye exhibits a distinct photophysical change. Binding of the dye to HEWL fibrils strongly suppresses the red AIE and enhances ICT emission. This is evidenced by a gradual decrease in the AIE intensity (∼660 nm) and an increase in LE (∼490 nm) and ICT (∼540 nm) emission intensities during the later stages of protein aggregation. Thus, the dye provides simultaneous measurements of the population dynamics of both HEWL oligomers and fibrils during protein aggregation based on the discriminatory modulation of AIE and ICT of the dye. The dye also enables imaging of both HEWL oligomers and fibrils simultaneously using different emission channels in super-resolution confocal fluorescence microscopy.
Collapse
Affiliation(s)
- Dharini Arumugam
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| | - Nidhi Anilkumar Jamuna
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| | - Adithya Kamalakshan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| | - Sarthak Mandal
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| |
Collapse
|
11
|
Streiber AM, Neitzel J, Nguyen Ho PT, Vernooij MW, Bos D. Intracranial arteriosclerosis is not associated with cerebral amyloid deposition. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70005. [PMID: 39360005 PMCID: PMC11444050 DOI: 10.1002/dad2.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/18/2024] [Accepted: 08/18/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Intracranial arteriosclerosis and cerebral amyloid beta (Aβ) are both involved in the etiology of Alzheimer's disease (AD) dementia, but the direct link between these two pathologies remains elusive. METHODS In 633 participants (mean age 69 years, 51% women) from the population-based Rotterdam Study, we quantified cerebral Aβ accumulation on amyloid positron emission tomography (PET). We assessed calcification of the intracranial internal carotid (ICAC) and vertebrobasilar arteries (VBAC) as proxies of arteriosclerosis on non-enhanced computed tomography (CT). Using logistic and linear regression, we studied the relationship of presence, burden, and type of calcification with the presence and burden of Aβ. RESULTS We found no associations of ICAC [odds ratio (OR): 0.85, 95% confidence interval (CI): 0.43, 1.72] or VBAC [OR: 0.59, CI: 0.26, 1.24] with cerebral Aβ. The results did not vary across ICAC subtypes. DISCUSSION Intracranial arteriosclerosis was not associated with cerebral Aβ, underscoring their independence in the etiology of AD dementia. Highlights Comprehensive assessment of intracranial arteriosclerosis (e.g., including subtypes).Intracranial arteriosclerosis in different arteries and cerebral Aβ are not related.Arteriosclerosis and Aβ likely influence Alzheimer's disease dementia independently.
Collapse
Affiliation(s)
- Anna M Streiber
- Department of Radiology and Nuclear Medicine Erasmus MC Rotterdam the Netherlands
- Department of Epidemiology Erasmus MC Rotterdam the Netherlands
| | - Julia Neitzel
- Department of Radiology and Nuclear Medicine Erasmus MC Rotterdam the Netherlands
- Department of Epidemiology Erasmus MC Rotterdam the Netherlands
- Department of Epidemiology Harvard T.H. Chan School of Public Health Boston Massachusetts USA
| | | | - Meike W Vernooij
- Department of Radiology and Nuclear Medicine Erasmus MC Rotterdam the Netherlands
- Department of Epidemiology Erasmus MC Rotterdam the Netherlands
| | - Daniel Bos
- Department of Radiology and Nuclear Medicine Erasmus MC Rotterdam the Netherlands
- Department of Epidemiology Erasmus MC Rotterdam the Netherlands
- Department of Epidemiology Harvard T.H. Chan School of Public Health Boston Massachusetts USA
| |
Collapse
|
12
|
Yang L, Gan YH, He XY, Deng YT, Zhang W, You J, Kuo K, Zhang YR, Huang SY, Wu BS, Guo Y, Zhang Y, Dong Q, Feng JF, Cheng W, Yu JT. Shift work effects on incident neuropsychiatric disorders and shift work tolerance. J Affect Disord 2024; 362:323-333. [PMID: 38971194 DOI: 10.1016/j.jad.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 02/28/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Shift work is associated with susceptibility to several neuropsychiatric disorders. This study aims to investigate the effect of shift work on the incidence of neuropsychiatric disorders, and highlighting how individual variability may influence the association. METHODS UK Biobank participants with employment information were included. Cox survival was conducted in main and subgroup analyses. Correlation analyses explored the impact of shift work on brain structures, and mediation analyses were performed to elucidate the shared underlying mechanisms. Shift work tolerance was evaluated through survival analyses contrasting the risks associated with five neuropsychiatric disorders in shift versus non-shift workers across different demographic or occupational strata. RESULTS The analysis encompassed 254,646 participants. Shift work was associated with higher risk of dementia (HR 1.29, 95 % CI 1.10-1.52), anxiety (1.08, 1.01-1.15), depression (1.29, 1.22-1.36), and sleep disorders (1.18, 1.09-1.28), but not stroke (p = 0.20). Shift work was correlated with decreasing volume of various brain regions, particularly in thalamus, lateral orbitofrontal, and middle temporal. Mediation analysis revealed that increased immune response and glucose levels are common pathways linking shift work to these disorders. We observed diversity in shift work tolerance across different individual characteristics, among which socioeconomic status and length of working hours were the most essential. LIMITATIONS Self-reported employment information may cause misclassification and recall bias. And since we focused on the middle-aged population, the conclusions may not be representative of younger or older populations. CONCLUSIONS Our findings indicated the need to monitor shift worker health and provide personalized management to help adapt to shift work.
Collapse
Affiliation(s)
- Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Han Gan
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Jia You
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Kevin Kuo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Wei Cheng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
13
|
Makhkamov M, Baev A, Kurganov E, Razzokov J. Understanding Osaka mutation polymorphic Aβ fibril response to static and oscillating electric fields: insights from computational modeling. Sci Rep 2024; 14:22246. [PMID: 39333193 PMCID: PMC11436846 DOI: 10.1038/s41598-024-72778-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder, impacting millions of individuals worldwide. Among its defining characteristics is the accumulation of senile plaques within the brain's gray matter, formed through the self-assembly of misfolded proteins contributing to the progressive symptoms of AD. This study investigates a polymorphic Aβ fibril under static and oscillating electric fields using molecular dynamics simulation. Specifically, we utilized a polymorphic fibrillar complex composed of two intertwined pentamer-strands of the Aβ1-40 peptide with the Osaka mutation (E22Δ), known for its toxicity and stable structure. Our findings demonstrate that a 0.3 and 0.4 V/nm electric field combined with a 0.20 GHz frequency effectively disrupts the polymorphic conformation of Aβ fibrils. Furthermore, we elucidate the molecular mechanisms underlying this disruption, providing insights into the potential therapeutic use of oscillating electric fields for AD. This research offers valuable insights into novel therapeutic approaches for combating AD pathology.
Collapse
Affiliation(s)
- Mukhriddin Makhkamov
- Laboratory of Experimental Biophysics, Centre for Advanced Technologies, Universitet 7, 100174, Tashkent, Uzbekistan
- Department of Information Technologies, Tashkent International University of Education, Imom Bukhoriy 6, 100207, Tashkent, Uzbekistan
| | - Artyom Baev
- Laboratory of Experimental Biophysics, Centre for Advanced Technologies, Universitet 7, 100174, Tashkent, Uzbekistan
| | - Erkin Kurganov
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Jamoliddin Razzokov
- Institute of Fundamental and Applied Research, National Research University TIIAME, Kori Niyoziy 39, 100000, Tashkent, Uzbekistan.
- Department of Biotechnology, School of Engineering, Tashkent State Technical University, 100095, Tashkent, Uzbekistan.
- Department of Natural Sciences, Shakhrisabz State Pedagogical Institute, Shakhrisabz Street 10, 181301, Kashkadarya, Uzbekistan.
| |
Collapse
|
14
|
Yelamanda Rao K, Chandran R, Dileep KV, Gorantla SC, Jeelan Basha S, Mothukuru S, Siva Kumar I, Vamsi K, Kumar S, Reddy ABM, Subramanyam R, Damu AG. Quinazolinone-Hydrazine Cyanoacetamide Hybrids as Potent Multitarget-Directed Druggable Therapeutics against Alzheimer's Disease: Design, Synthesis, and Biochemical, In Silico, and Mechanistic Analyses. ACS Chem Neurosci 2024; 15:3401-3420. [PMID: 39235838 DOI: 10.1021/acschemneuro.4c00424] [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: 09/06/2024] Open
Abstract
The discovery of effective multitarget-directed ligands (MTDLs) against multifactorial Alzheimer's disease (AD) remnants has been focused in an incessant drug discovery pursuit. In this perception, the current study explores the rational design, synthesis, and evaluation of 26 quinazolinone-hydrazine cyanoacetamide hybrids 7(a-j), 8(a-j), and 9(a-f) as MTDLs against AD. These new compounds were synthesized in four-step processes using simple phthalimide as the starting material without any major workup procedures and were characterized by different spectroscopic techniques. In Ellman's assay, the most potent analogues 7i, 8j, and 9d were identified as selective and mixed-type inhibitors of hAChE. Furthermore, biophysical and computational assessments revealed that the analogues 7i, 8j, and 9d were bound to both the catalytic active site and peripheral anionic site of hAChE with high affinity. The molecular dynamics simulation analysis highlighted the conformational changes of hAChE upon binding of 7i, 8j, and 9d and also the stability of resulting biomolecular systems all over 100 ns simulations. In addition to antioxidant activity, the most active congeners were found to protect substantially SK-N-SH cells from oxidative damage. Decisively, the most active analogues 7i, 8j, and 9d were assessed as potent Aβ1-42 fibril modulators and protective agents against Aβ1-42-induced toxicity in SH-SY5Y cells. Additionally, glioblastoma C6 cell-based assays also demonstrated the use of the most active congeners 7i, 8j, and 9d as protective agents against Aβ1-42-induced toxicity. Overall, this multifunctional capacity of quinazolinone-hydrazine cyanoacetamide hybrids demonstrated the noteworthy potential of these hybrids to develop as effectual MTDLs against AD. However, further pharmacokinetics, toxicology, and behavioral studies are warranted.
Collapse
Affiliation(s)
- Kandrakonda Yelamanda Rao
- Bioorganic Chemistry Research Laboratory, Department of Chemistry, Yogi Vemana University, Kadapa, Andhra Pradesh 516005, India
| | - Remya Chandran
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala 680005, India
| | - K V Dileep
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala 680005, India
| | - Sri Charitha Gorantla
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Shaik Jeelan Basha
- Bioorganic Chemistry Research Laboratory, Department of Chemistry, Yogi Vemana University, Kadapa, Andhra Pradesh 516005, India
- Department of Chemistry, Santhiram Engineering College (Autonomous), Nandyal, Andhra Pradesh 518501, India
| | - Sreelakshmi Mothukuru
- Bioorganic Chemistry Research Laboratory, Department of Chemistry, Yogi Vemana University, Kadapa, Andhra Pradesh 516005, India
| | - Irla Siva Kumar
- Soft Condensed Matter, Raman Research Institute, CV Raman Avenue, Sadashiva Nagar, Bangalore 560080, India
| | - Katta Vamsi
- Department of Chemistry, Indian Institute of Science and Education Research (IISER), Tirupati, Andhra Pradesh 517507, India
| | - Sandeep Kumar
- Soft Condensed Matter, Raman Research Institute, CV Raman Avenue, Sadashiva Nagar, Bangalore 560080, India
- Nitte Meenakshi Institute of Technology, Yelahanka, Bangalore 560064, India
| | - Aramati Bindu Madhava Reddy
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Rajagopal Subramanyam
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana 500046, India
| | - Amooru Gangaiah Damu
- Bioorganic Chemistry Research Laboratory, Department of Chemistry, Yogi Vemana University, Kadapa, Andhra Pradesh 516005, India
| |
Collapse
|
15
|
Kale MB, Bhondge HM, Wankhede NL, Shende PV, Thanekaer RP, Aglawe MM, Rahangdale SR, Taksande BG, Pandit SB, Upaganlawar AB, Umekar MJ, Kopalli SR, Koppula S. Navigating the intersection: Diabetes and Alzheimer's intertwined relationship. Ageing Res Rev 2024; 100:102415. [PMID: 39002642 DOI: 10.1016/j.arr.2024.102415] [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: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Alzheimer's disease (AD) and Diabetes mellitus (DM) exhibit comparable pathophysiological pathways. Genetic abnormalities in APP, PS-1, and PS-2 are linked to AD, with diagnostic aid from CSF and blood biomarkers. Insulin dysfunction, termed "type 3 diabetes mellitus" in AD, involves altered insulin signalling and neuronal shrinkage. Insulin influences beta-amyloid metabolism, exacerbating neurotoxicity in AD and amyloid production in DM. Both disorders display impaired glucose transporter expression, hastening cognitive decline. Mitochondrial dysfunction and Toll-like receptor 4-mediated inflammation worsen neurodegeneration in both diseases. ApoE4 raises disease risk, especially when coupled with dyslipidemia common in DM. Targeting shared pathways like insulin-degrading enzyme activation and HSP60 holds promise for therapeutic intervention. Recognizing these interconnected mechanisms underscores the imperative for developing tailored treatments addressing the overlapping pathophysiology of AD and DM, offering potential avenues for more effective management of both conditions.
Collapse
Affiliation(s)
- Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | | | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Rushikesh P Thanekaer
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Manish M Aglawe
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sandip R Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Sunil B Pandit
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|
16
|
McDade E, Liu H, Bui Q, Hassenstab J, Gordon B, Benzinger T, Shen Y, Timsina J, Wang L, Sung YJ, Karch C, Renton A, Daniels A, Morris J, Xiong C, Ibanez L, Perrin R, Llibre-Guerra JJ, Day G, Supnet-Bell C, Xu X, Berman S, Chhatwal J, Ikeuchi T, Kasuga K, Niimi Y, Huey E, Schofield P, Brooks W, Ryan N, Jucker M, Laske C, Levin J, Vöglein J, Roh JH, Lopera F, Bateman R, Cruchaga C. Ubiquitin-Proteasome System in the Different Stages of Dominantly Inherited Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-4202125. [PMID: 39108475 PMCID: PMC11302696 DOI: 10.21203/rs.3.rs-4202125/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
This study explored the role of the ubiquitin-proteasome system (UPS) in dominantly inherited Alzheimer's disease (DIAD) by examining changes in cerebrospinal fluid (CSF) levels of UPS proteins along with disease progression, AD imaging biomarkers (PiB PET, tau PET), neurodegeneration imaging measures (MRI, FDG PET), and Clinical Dementia Rating® (CDR®). Using the SOMAscan assay, we detected subtle increases in specific ubiquitin enzymes associated with proteostasis in mutation carriers (MCs) up to two decades before the estimated symptom onset. This was followed by more pronounced elevations of UPS-activating enzymes, including E2 and E3 proteins, and ubiquitin-related modifiers. Our findings also demonstrated consistent correlations between UPS proteins and CSF biomarkers such as Aβ42/40 ratio, total tau, various phosphorylated tau species to total tau ratios (ptau181/T181, ptauT205/T205, ptauS202/S202, ptauT217/T217), and MTBR-tau243, alongside Neurofilament light chain (NfL) and the CDR®. Notably, a positive association was observed with imaging markers (PiB PET, tau PET) and a negative correlation with markers of neurodegeneration (FDG PET, MRI), highlighting a significant link between UPS dysregulation and neurodegenerative processes. The correlations suggest that the increase in multiple UPS proteins with rising tau levels and tau-tangle associated markers, indicating a potential role for the UPS in relation to misfolded tau/neurofibrillary tangles (NFTs) and symptom onset. These findings indicate that elevated CSF UPS proteins in DIAD MCs could serve as early indicators of disease progression and suggest a link between UPS dysregulation and amyloid plaque, tau tangles formation, implicating the UPS as a potential therapeutic target in AD pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Alan Renton
- Nash Family Department of Neuroscience and Ronald Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA: Departments of Neurology and Genetics and Ge
| | | | | | | | | | | | | | | | | | | | | | - Jasmeer Chhatwal
- Massachusetts General Hospital, Brigham and Women's Hospital, Harvard Medical School
| | | | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University
| | | | | | | | | | | | | | | | | | | | | | | | - Randall Bateman
- Department of Neurology, Washington University School of Medicine
| | | |
Collapse
|
17
|
Babalola JA, Stracke A, Loeffler T, Schilcher I, Sideromenos S, Flunkert S, Neddens J, Lignell A, Prokesch M, Pazenboeck U, Strobl H, Tadic J, Leitinger G, Lass A, Hutter-Paier B, Hoefler G. Effect of astaxanthin in type-2 diabetes -induced APPxhQC transgenic and NTG mice. Mol Metab 2024; 85:101959. [PMID: 38763496 PMCID: PMC11153249 DOI: 10.1016/j.molmet.2024.101959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVES Aggregation and misfolding of amyloid beta (Aβ) and tau proteins, suggested to arise from post-translational modification processes, are thought to be the main cause of Alzheimer's disease (AD). Additionally, a plethora of evidence exists that links metabolic dysfunctions such as obesity, type 2 diabetes (T2D), and dyslipidemia to the pathogenesis of AD. We thus investigated the combinatory effect of T2D and human glutaminyl cyclase activity (pyroglutamylation), on the pathology of AD and whether astaxanthin (ASX) treatment ameliorates accompanying pathophysiological manifestations. METHODS Male transgenic AD mice, APPxhQC, expressing human APP751 with the Swedish and the London mutation and human glutaminyl cyclase (hQC) enzyme and their non-transgenic (NTG) littermates were used. Both APPxhQC and NTG mice were allocated to 3 groups, control, T2D-control, and T2D-ASX. Mice were fed control or high fat diet ± ASX for 13 weeks starting at an age of 11-12 months. High fat diet fed mice were further treated with streptozocin for T2D induction. Effects of genotype, T2D induction, and ASX treatment were evaluated by analysing glycemic readouts, lipid concentration, Aβ deposition, hippocampus-dependent cognitive function and nutrient sensing using immunosorbent assay, ELISA-based assays, western blotting, immunofluorescence staining, and behavioral testing via Morris water maze (MWM), respectively. RESULTS APPxhQC mice presented a higher glucose sensitivity compared to NTG mice. T2D-induced brain dysfunction was more severe in NTG compared to the APPxhQC mice. T2D induction impaired memory functions while increasing hepatic LC3B, ABCA1, and p65 levels in NTG mice. T2D induction resulted in a progressive shift of Aβ from the soluble to insoluble form in APPxhQC mice. ASX treatment reversed T2D-induced memory dysfunction in NTG mice and in parallel increased hepatic pAKT while decreasing p65 and increasing cerebral p-S6rp and p65 levels. ASX treatment reduced soluble Aβ38 and Aβ40 and insoluble Aβ40 levels in T2D-induced APPxhQC mice. CONCLUSIONS We demonstrate that T2D induction in APPxhQC mice poses additional risk for AD pathology as seen by increased Aβ deposition. Although ASX treatment reduced Aβ expression in T2D-induced APPxhQC mice and rescued T2D-induced memory impairment in NTG mice, ASX treatment alone may not be effective in cases of T2D comorbidity and AD.
Collapse
Affiliation(s)
| | - Anika Stracke
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Austria
| | | | | | - Spyridon Sideromenos
- QPS Austria GmbH, Grambach, Austria; Medical University of Vienna, Vienna, Austria
| | | | | | | | | | - Ute Pazenboeck
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Herbert Strobl
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Jelena Tadic
- Institute of Molecular Biosciences, University of Graz, Austria
| | - Gerd Leitinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Austria
| | | | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology Medical University of Graz, Graz, Austria.
| |
Collapse
|
18
|
Thiankhaw K, Chattipakorn N, Chattipakorn SC. How calcineurin inhibitors affect cognition. Acta Physiol (Oxf) 2024; 240:e14161. [PMID: 38747643 DOI: 10.1111/apha.14161] [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: 11/18/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
AIMS With a focus on the discrepancy between preclinical and clinical findings, this review will gather comprehensive information about the effects of calcineurin inhibitors (CNI) on cognitive function and related brain pathology from in vitro, in vivo, and clinical studies. We also summarize the potential mechanisms that underlie the pathways related to CNI-induced cognitive impairment. METHODS We systematically searched articles in PubMed using keywords 'calcineurin inhibitor*' and 'cognition' to identify related articles, which the final list pertaining to underlying mechanisms of CNI on cognition. RESULTS Several studies have reported an association between calcineurin and the neuropathology of Alzheimer's disease (AD). AD is the most common neurocognitive disorder associated with amyloid plaques and neurofibrillary tangles in the brain, leading to cognitive impairment. CNI, including tacrolimus and cyclosporin A, are commonly prescribed for patients with transplantation of solid organs such as kidney, liver, or heart, those drugs are currently being used as long-term immunosuppressive therapy. Although preclinical models emphasize the favorable effects of CNI on the restoration of brain pathology due to the impacts of calcineurin on the alleviation of amyloid-beta deposition and tau hyperphosphorylation, or rescuing synaptic and mitochondrial functions, treatment-related neurotoxicity, resulting in cognitive dysfunctions has been observed in clinical settings of patients who received CNI. CONCLUSION Inconsistent results of CNI on cognition from clinical studies have been observed due to impairment of the blood-brain barrier, neuroinflammation mediated by reactive oxygen species, and alteration in mitochondrial fission, and extended research is required to confirm its promising use in cognitive impairment.
Collapse
Affiliation(s)
- Kitti Thiankhaw
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siripron C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
19
|
Chen Y, Huang X, Chen H, Yi C. An easy-to-perform method for microvessel isolation and primary brain endothelial cell culture to study Alzheimer's disease. Heliyon 2024; 10:e33077. [PMID: 38994107 PMCID: PMC11238044 DOI: 10.1016/j.heliyon.2024.e33077] [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: 04/09/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Dysfunction of the blood-brain barrier (BBB) has been increasingly recognised as a critical early event in Alzheimer's disease (AD) pathophysiology. Central to this mechanism is the impaired function of brain endothelial cells (BECs), the primary structural constituents of the BBB, the study of which is imperative for understanding AD pathophysiology. However, the published methods to isolate BECs are time-consuming and have a low success rate. Here, we developed a rapid and streamlined protocol for BEC isolation without using transgenic reporters, flow cytometry, and magnetic beads, which are essential for existing methods. Using this novel protocol, we isolated high-purity BECs from cell clusters of cortical microvessels from wild-type and APPswe/PS1dE9 (APP/PS1, a classical AD model) mice at 2, 4 and 9 months of age. Reduced levels of tight junction proteins Claudin-5 and Zonula Occludens-1, as well as glucose transporter 1, were observed in the isolated cortical microvessels from APP/PS1 mice and amyloid-β (Aβ) oligomer-treated BECs from wild-type mice. Trans-well permeability assay showed increased FITC-dextran leakage in BECs treated with Aβ, suggesting impaired BBB permeability. BECs obtained using our novel protocol can undergo various experimental analyses, including immunofluorescence staining, western blotting, real-time PCR, and trans-well permeability assay. In conclusion, our novel protocol represents a reliable and valuable tool for in vitro modelling BBB to study AD-related mechanisms and develop targeted therapeutic strategies.
Collapse
Affiliation(s)
- Yang Chen
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiaomin Huang
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, 518107, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China
| |
Collapse
|
20
|
Kalluru PKR, Bhavanthi S, Vashist S, Gopavaram RR, Mamilla M, Sharma S, Gundoji CR, Goguri SR. Role of erythropoietin in the treatment of Alzheimer's disease: the story so far. Ann Med Surg (Lond) 2024; 86:3608-3614. [PMID: 38846819 PMCID: PMC11152865 DOI: 10.1097/ms9.0000000000002113] [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: 01/20/2024] [Accepted: 04/17/2024] [Indexed: 06/09/2024] Open
Abstract
This review aims to explore the potential of erythropoietin, a glycopeptide hormone, as a treatment option for Alzheimer's disease, which is the commonest cause of dementia. Despite years of focus and research, therapeutic options for Alzheimer's disease are not yet completely satisfactory. And as people age, they are likely to develop Alzheimer's Disease, further pressuring the healthcare system. So, it is definite to develop treatment options that meet superior outcomes with minimal negative effects. A comprehensive review of the literature was conducted in PubMed and Google Scholar using a combination of keywords, including Alzheimer's disease, dementia, erythropoietin, and neuroprotection. Search results were assessed for relevance before using the data for this study. The beneficial implications of erythropoietin as a therapeutic option have been explored, along with the side effects and mechanisms of erythropoietin in Alzheimer's disease. Overall, the authors' review indicates that erythropoietin presents a promising avenue for mitigating the progression of Alzheimer's disease, with minimal associated side effects.
Collapse
Affiliation(s)
| | | | | | | | | | - Shriya Sharma
- Internal Medicine, Dnipropetrovsk Medical Academy of Health Ministry of Ukraine, Dnipro, Ukraine
| | | | | |
Collapse
|
21
|
Thai QM, Tran PT, Phung HTT, Pham MQ, Ngo ST. Silver nanoparticles alter the dimerization of Aβ 42 studied by REMD simulations. RSC Adv 2024; 14:15112-15119. [PMID: 38720971 PMCID: PMC11078207 DOI: 10.1039/d4ra02197e] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/03/2024] [Indexed: 01/06/2025] Open
Abstract
The aggregation of amyloid beta (Aβ) peptides is associated with the development of Alzheimer's disease (AD). However, there has been a growing belief that the oligomerization of Aβ species in different environments has a neurotoxic effect on the patient's brain, causing damage. It is necessary to comprehend the compositions of Aβ oligomers in order to develop medications that may effectively inhibit these neurotoxic forms that affect the nervous system of AD patients. Thus, dissociation or inhibition of Aβ aggregation may be able to prevent AD. To date, the search for traditional agents and biomolecules has largely been unsuccessful. In this context, nanoparticles have emerged as potential candidates to directly inhibit the formation of Aβ oligomers. The oligomerization of the dimeric Aβ peptides with or without the influence of a silver nanoparticle was thus investigated using temperature replica-exchange molecular dynamics (REMD) simulations. The physical insights into the dimeric Aβ oligomerization were clarified by analyzing intermolecular contact maps, the free energy landscape of the dimeric oligomer, secondary structure terms, etc. The difference in obtained metrics between Aβ with or without a silver nanoparticle provides a picture of the influence of silver nanoparticles on the oligomerization process. The underlying mechanisms that are involved in altering Aβ oligomerization will be discussed. The obtained results may play an important role in searching for Aβ inhibitor pathways.
Collapse
Affiliation(s)
- Quynh Mai Thai
- Laboratory of Biophysics, Institute of Advanced Study in Technology, Ton Duc Thang University Ho Chi Minh City Vietnam
- Faculty of Pharmacy, Ton Duc Thang University Ho Chi Minh City Vietnam
| | | | - Huong T T Phung
- NTT Hi-Tech Institute, Nguyen Tat Thanh University Ho Chi Minh City Vietnam
| | - Minh Quan Pham
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology Hanoi Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology Hanoi Vietnam
| | - Son Tung Ngo
- Laboratory of Biophysics, Institute of Advanced Study in Technology, Ton Duc Thang University Ho Chi Minh City Vietnam
- Faculty of Pharmacy, Ton Duc Thang University Ho Chi Minh City Vietnam
| |
Collapse
|
22
|
Wongchitrat P, Chanmee T, Govitrapong P. Molecular Mechanisms Associated with Neurodegeneration of Neurotropic Viral Infection. Mol Neurobiol 2024; 61:2881-2903. [PMID: 37946006 PMCID: PMC11043213 DOI: 10.1007/s12035-023-03761-6] [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: 08/25/2022] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Viral infections of the central nervous system (CNS) cause variable outcomes from acute to severe neurological sequelae with increased morbidity and mortality. Viral neuroinvasion directly or indirectly induces encephalitis via dysregulation of the immune response and contributes to the alteration of neuronal function and the degeneration of neuronal cells. This review provides an overview of the cellular and molecular mechanisms of virus-induced neurodegeneration. Neurotropic viral infections influence many aspects of neuronal dysfunction, including promoting chronic inflammation, inducing cellular oxidative stress, impairing mitophagy, encountering mitochondrial dynamics, enhancing metabolic rewiring, altering neurotransmitter systems, and inducing misfolded and aggregated pathological proteins associated with neurodegenerative diseases. These pathogenetic mechanisms create a multidimensional injury of the brain that leads to specific neuronal and brain dysfunction. The understanding of the molecular mechanisms underlying the neurophathogenesis associated with neurodegeneration of viral infection may emphasize the strategies for prevention, protection, and treatment of virus infection of the CNS.
Collapse
Affiliation(s)
- Prapimpun Wongchitrat
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon 4 Road, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand.
| | - Theerawut Chanmee
- Department of Clinical Chemistry, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | | |
Collapse
|
23
|
Lee D, Antonsdottir IM, Clark ED, Porsteinsson AP. Review of valiltramiprosate (ALZ-801) for the treatment of Alzheimer's disease: a novel small molecule with disease modifying potential. Expert Opin Pharmacother 2024; 25:791-799. [PMID: 38814590 DOI: 10.1080/14656566.2024.2360069] [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: 03/31/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative condition characterized by progressive cognitive deterioration, functional impairments, and neuropsychiatric symptoms. Valiltramiprosate is a tramiprosate prodrug being investigated as a novel treatment for AD. AREAS COVERED The online databases PubMed, Embase, Web of Science, Cochrane Library, and ClinicalTrials.gov were searched using the terms 'ALZ-801' or 'valiltramiprosate.' Alzheon press releases were reviewed for emerging clinical information. Valiltramiprosate is an oral, well-tolerated synthetic valine-conjugate prodrug of tramiprosate. Valiltramiprosate's active metabolite include tramiprosate and 3-sulfopropanoic acid. Proposed mechanism of action is multiligand binding to Aβ42 which stabilizes amyloid monomers to prevent peptide aggregation and oligomerization. Pharmacokinetic studies show 52% oral bioavailability, rapid absorption, approximately 40% brain-drug exposure, and near complete renal clearance. Compared to tramiprosate, valiltramiprosate extends plasma tramiprosate half-life and improves interindividual pharmacokinetic variability. Interim analyses from valiltramiprosate's phase II biomarker trial show: (1) significant reductions in plasma p-tau181 and related AD fluid biomarkers; (2) brain structure preservation and reduced hippocampal atrophy by MRI; and (3) improvements on cognitive assessments at multiple timepoints. Its phase III clinical trial in ApoE ε4 homozygotes is near completion. EXPERT OPINION Valiltramiprosate's clinical trial data show early indications of efficacy with potential disease modifying effect in AD.
Collapse
Affiliation(s)
- Daniel Lee
- Alzheimer's Disease Care, Research and Education (AD-CARE), Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Inga M Antonsdottir
- Johns Hopkins School of Nursing, Baltimore, MD, USA
- Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Department of Psychiatry and Behavioral Sciences, Johns Hopkins Bayview, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Emily D Clark
- Alzheimer's Disease Care, Research and Education (AD-CARE), Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Anton P Porsteinsson
- Alzheimer's Disease Care, Research and Education (AD-CARE), Department of Psychiatry, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
24
|
Khan A, Untoo R, Patthi B, Singla A, Kumari M, Rajeev A. Oral Health-Related Quality of Life (OHRQoL) in Elderly Patients with and without Alzheimer's Disease - An Evaluative Study. Indian J Dent Res 2024; 35:149-153. [PMID: 39282766 DOI: 10.4103/ijdr.ijdr_792_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 03/01/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterised by the loss of intellectual functions. The disorder is usually seen after the age of 60 years, and these elderly patients tend to experience a progressive decline in their cognitive and behavioural abilities. AIM The present study was carried out to assess the oral health-related quality of life (OHRQoL) in elderly patients suffering from AD as compared to subjects without the disease. MATERIALS AND METHOD One hundred elderly subjects with AD and 150 subjects without AD were included in the study. The subjective assessment of OHRQoL was done using the General Oral Health Assessment Index (GOHAI). Demographic and oral characteristics were assessed for dentition status with the Decayed, Missing and Filled Teeth (DMFT) index, Oral Hygiene Index (OHI), prosthesis use, xerostomia and presence of oral pathologies. RESULTS GOHAI values were found to be statistically higher ( P = 0.0003) in the subjects with AD. The subjects with AD were found to have a higher DMFT (mean difference = 4.11), Oral Hygiene Index (OHI) scores (mean difference = 1.54), xerostomia ( P = 0.0128) and number of oral pathologies. However, the educational level ( P = 0.2603) and number of natural teeth ( P = 0.0006) were found to be lower in the group with AD. CONCLUSION The present study reported that the elderly subjects suffering from AD had poorer OHRQoL than those without the disease. There is a need to comprehensively evaluate the oral health conditions of elderly patients with AD and actively provide management to improve their OHRQoL.
Collapse
Affiliation(s)
- Ambar Khan
- Department of Public Health Dentistry, College of Dental Science and Hospital, Indore, Madhya Pradesh, India
| | - Rafia Untoo
- Department of Public Health Dentistry, D.J. College of Dental Sciences and Research, Modinagar, Uttar Pradesh, India
| | - Basavaraj Patthi
- Department of Public Health Dentistry, D.J. College of Dental Sciences and Research, Modinagar, Uttar Pradesh, India
| | - Ashish Singla
- Department of Public Health Dentistry, D.J. College of Dental Sciences and Research, Modinagar, Uttar Pradesh, India
| | - Monika Kumari
- Department of Public Health Dentistry, Dental College Azamgarh, Uttar Pradesh, India
| | - Ananthalekshmy Rajeev
- Department of Public Health Dentistry, The Oxford Dental College, Bengaluru, Karnataka, India
| |
Collapse
|
25
|
Mrđenović D, Combes BF, Ni R, Zenobi R, Kumar N. Probing Chemical Complexity of Amyloid Plaques in Alzheimer's Disease Mice using Hyperspectral Raman Imaging. ACS Chem Neurosci 2024; 15:78-85. [PMID: 38096362 PMCID: PMC10767745 DOI: 10.1021/acschemneuro.3c00607] [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: 09/18/2023] [Revised: 11/11/2023] [Accepted: 11/30/2023] [Indexed: 01/04/2024] Open
Abstract
One of the distinctive pathological features of Alzheimer's disease (AD) is the deposition of amyloid plaques within the brain of affected individuals. These plaques have traditionally been investigated using labeling techniques such as immunohistochemical imaging. However, the use of labeling can disrupt the structural integrity of the molecules being analyzed. Hence, it is imperative to employ label-free imaging methods for noninvasive examination of amyloid deposits in their native form, thereby providing more relevant information pertaining to AD. This study presents compelling evidence that label-free and nondestructive confocal Raman imaging is a highly effective approach for the identification and chemical characterization of amyloid plaques within cortical regions of an arcAβ mouse model of AD. Furthermore, this investigation elucidates how the spatial correlation of Raman signals can be exploited to identify robust Raman marker bands and discern proteins and lipids from amyloid plaques. Finally, this study uncovers the existence of distinct types of amyloid plaques in the arcAβ mouse brain, exhibiting significant disparities in terms of not only shape and size but also molecular composition.
Collapse
Affiliation(s)
- Dušan Mrđenović
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 1−5/10, 8093 Zürich, Switzerland
| | - Benjamin F. Combes
- Institute
for Regenerative Medicine, University of
Zürich, Wagistrasse
12, 8952 Schlieren, Switzerland
| | - Ruiqing Ni
- Institute
for Regenerative Medicine, University of
Zürich, Wagistrasse
12, 8952 Schlieren, Switzerland
- Institute
for Biomedical Engineering, University of
Zurich and ETH Zurich, Wolfgang-Pauli-Strasse 27, 8093 Zürich, Switzerland
| | - Renato Zenobi
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 1−5/10, 8093 Zürich, Switzerland
| | - Naresh Kumar
- Department
of Chemistry and Applied Biosciences, ETH
Zürich, Vladimir-Prelog-Weg 1−5/10, 8093 Zürich, Switzerland
| |
Collapse
|
26
|
Zhang M, Luo X, Zhang B, Luo D, Huang L, Long Q. Unveiling OSCP as the potential therapeutic target for mitochondrial dysfunction-related diseases. Life Sci 2024; 336:122293. [PMID: 38030056 DOI: 10.1016/j.lfs.2023.122293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
Mitochondria are important organelles in cells responsible for energy production and regulation. Mitochondrial dysfunction has been implicated in the pathogenesis of many diseases. Oligomycin sensitivity-conferring protein (OSCP), a component of the inner mitochondrial membrane, has been studied for a long time. OSCP is a component of the F1Fo-ATP synthase in mitochondria and is closely related to the regulation of the mitochondrial permeability transition pore (mPTP). Studies have shown that OSCP plays an important role in cardiovascular disease, neurological disorders, and tumor development. This review summarizes the localization, structure, function, and regulatory mechanisms of OSCP and outlines its role in cardiovascular disease, neurological disease, and tumor development. In addition, this article reviews the research on the interaction between OSCP and mPTP. Finally, the article suggests future research directions, including further exploration of the mechanism of action of OSCP, the interaction between OSCP and other proteins and signaling pathways, and the development of new treatment strategies for mitochondrial dysfunction. In conclusion, in-depth research on OSCP will help to elucidate its importance in cell function and disease and provide new ideas for the treatment and prevention of related diseases.
Collapse
Affiliation(s)
- Mingyue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xia Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Binzhi Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Duosheng Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lizhen Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qinqiang Long
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
27
|
Zhang X, Haeri M, Swerdlow RH, Wang N. Loss of Adaptive DNA Breaks in Alzheimer's Disease Brains. J Alzheimers Dis 2024; 97:1861-1875. [PMID: 38306051 PMCID: PMC10894583 DOI: 10.3233/jad-231303] [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] [Accepted: 12/16/2023] [Indexed: 02/03/2024]
Abstract
Background DNA breaks accumulate in Alzheimer's disease (AD) brains. While their role as true genomic lesions is recognized, DNA breaks also support cognitive function by facilitating the expression of activity-dependent immediate early genes. This process involves TOP2B, a DNA topoisomerase that catalyzes the formation of DNA double-strand breaks. Objective To characterize how AD impacts adaptive DNA breaks at nervous system genes. Methods We leveraged the ability of DNA single- and double-strand breaks to activate poly(ADP-ribose) polymerases (PARPs) that conjugate poly(ADP-ribose) (PAR) to adjacent proteins. To characterize the genomic sites harboring DNA breaks in AD brains, nuclei extracted from 3 AD and 3 non-demented autopsy brains (frontal cortex, all male donors, age 78 to 91 years of age) were analyzed through CUT&RUN in which we targeted PAR with subsequent DNA sequencing. Results Although the AD brains contained 19.9 times more PAR peaks than the non-demented brains, PAR peaks at nervous system genes were profoundly lost in AD brains, and the expression of these genes was downregulated. This result is consistent with our previous CUT&RUN targeting γH2AX, which marks DNA double-strand breaks. In addition, TOP2B expression was significantly decreased in the AD brains. Conclusions Although AD brains contain a net increase in DNA breaks, adaptive DNA breaks at nervous system genes are lost in AD brains. This could potentially reflect diminished TOP2B expression and contribute to impaired neuron function and cognition in AD patients.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Institute of Reproduction and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mohammad Haeri
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H. Swerdlow
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ning Wang
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Institute of Reproduction and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
28
|
Tufail S, Sherwani MA, Shamim Z, Abdullah, Goh KW, Alomary MN, Ansari MA, Almosa AA, Ming LC, Abdullah ADI, Khan FB, Menhali AA, Mirza S, Ayoub MA. 2D nanostructures: Potential in diagnosis and treatment of Alzheimer's disease. Biomed Pharmacother 2024; 170:116070. [PMID: 38163396 DOI: 10.1016/j.biopha.2023.116070] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/06/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
Two-dimensional (2D) nanomaterials have garnered enormous attention seemingly due to their unusual architecture and properties. Graphene and graphene oxide based 2D nanomaterials remained the most sought after for several years but the quest to design superior 2D nanomaterials which can find wider application gave rise to development of non-graphene 2D materials as well. Consequently, in addition to graphene based 2D nanomaterials, 2D nanostructures designed using macromolecules (such as DNAs, proteins, peptides and peptoids), transition metal dichalcogenides, transition-metal carbides and/or nitrides (MXene), black phosphorous, chitosan, hexagonal boron nitrides, and graphitic carbon nitride, and covalent organic frameworks have been developed. Interestingly, these 2D nanomaterials have found applications in diagnosis and treatment of various diseases including Alzheimer's disease (AD). Although AD is one of the most debilitating neurodegenerative conditions across the globe; unfortunately, there remains a paucity of effective diagnostic and/or therapeutic intervention for it till date. In this scenario, nanomaterial-based biosensors, or therapeutics especially 2D nanostructures are emerging to be promising in this regard. This review summarizes the diagnostic and therapeutic platforms developed for AD using 2D nanostructures. Collectively, it is worth mentioning that these 2D nanomaterials would seemingly provide an alternative and intriguing platform for biomedical interventions.
Collapse
Affiliation(s)
- Saba Tufail
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA.
| | | | - Zahid Shamim
- Department of Electrical Engineering, Faculty of Engineering and Technology, Aligarh Muslim University, Aligarh, India
| | - Abdullah
- Department of Pharmacy, University of Malakand, Khyber Pakhtunkhwa, Pakistan
| | - Khang Wen Goh
- Faculty Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
| | - Abdulaziz Abdullah Almosa
- Wellness and Preventive Medicine Institute, King AbdulAziz City of Science and Technology, Riyadh, Saudi Arabia.
| | - Long Chiau Ming
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia.
| | - Amar Daud Iskandar Abdullah
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia.
| | - Farheen Badrealam Khan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates; Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, PO Box 127788, Abu Dhabi, United Arab Emirates.
| | - Asma Al Menhali
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Sameer Mirza
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Mohammed Akli Ayoub
- Department of Biological Sciences, College of Medicine and Health Sciences, Khalifa University, PO Box 127788, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
29
|
Nguyen-Thi PT, Ho TT, Nguyen TT, Vo GV. Nanotechnology-based Drug Delivery for Alzheimer's and Parkinson's Diseases. Curr Drug Deliv 2024; 21:917-931. [PMID: 37424345 DOI: 10.2174/1567201820666230707113405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 05/03/2023] [Accepted: 05/12/2023] [Indexed: 07/11/2023]
Abstract
The delivery of drugs to the brain is quite challenging in the treatment of the central nervous system (CNS) diseases due to the blood-brain barrier and the blood-cerebrospinal fluid barrier. However, significant developments in nanomaterials employed by nanoparticle drug-delivery systems have substantial potential to cross or bypass these barriers leading to enhanced therapeutic efficacies. Advances in nanoplatform, nanosystems based on lipids, polymers and inorganic materials have been extensively studied and applied in treating Alzheimer's and Parkinson's diseases. In this review, various types of brain drug delivery nanocarriers are classified, summarized, and their potential as drug delivery systems in Alzheimer's and Parkinson's diseases is discussed. Finally, challenges facing the clinical translation of nanoparticles from bench to bedside are highlighted.
Collapse
Affiliation(s)
| | - Thanh-Tam Ho
- Institute for Global Health Innovations, Duy Tan University, Da Nang 550000, Vietnam
- Faculty of Pharmacy, Duy Tan University, Da Nang 550000, Vietnam
| | - Thuy Trang Nguyen
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City 71420, Vietnam
| | - Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University, Ho Chi Minh City [VNU-HCM], Ho Chi Minh City 700000, Vietnam
- Research Center for Genetics and Reproductive Health [CGRH], School of Medicine, Vietnam National University, Ho Chi Minh City [VNU-HCM], Ho Chi Minh City 70000, Vietnam
- Vietnam National University, Ho Chi Minh City [VNU-HCM], Ho Chi Minh City 700000, Vietnam
| |
Collapse
|
30
|
Marković M, Milošević J, Wang W, Cao Y. Passive Immunotherapies Targeting Amyloid- β in Alzheimer's Disease: A Quantitative Systems Pharmacology Perspective. Mol Pharmacol 2023; 105:1-13. [PMID: 37907353 DOI: 10.1124/molpharm.123.000726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/04/2023] [Accepted: 10/10/2023] [Indexed: 11/02/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by amyloid-β (Aβ) protein accumulation in the brain. Passive immunotherapies using monoclonal antibodies for targeting Aβ have shown promise for AD treatment. Indeed, recent US Food and Drug Administration approval of aducanumab and lecanemab, alongside positive donanemab Phase III results demonstrated clinical efficacy after decades of failed clinical trials for AD. However, the pharmacological basis distinguishing clinically effective from ineffective therapies remains unclear, impeding development of potent therapeutics. This study aimed to provide a quantitative perspective for effectively targeting Aβ with antibodies. We first reviewed the contradicting results associated with the amyloid hypothesis and the pharmacological basis of Aβ immunotherapy. Subsequently, we developed a quantitative systems pharmacology (QSP) model that describes the non-linear progression of Aβ pathology and the pharmacologic actions of the Aβ-targeting antibodies. Using the QSP model, we analyzed various scenarios for effective passive immunotherapy for AD. The model revealed that binding exclusively to the Aβ monomer has minimal effect on Aβ aggregation and plaque reduction, making the antibody affinity toward Aβ monomer unwanted, as it could become a distractive mechanism for plaque reduction. Neither early intervention, high brain penetration, nor increased dose could yield significant improvement of clinical efficacy for antibodies targeting solely monomers. Antibodies that bind all Aβ species but lack effector function exhibited moderate effects in plaque reduction. Our model highlights the importance of binding aggregate Aβ species and incorporating effector functions for efficient and early plaque reduction, guiding the development of more effective therapies for this devastating disease. SIGNIFICANCE STATEMENT: Despite previous unsuccessful attempts spanning several decades, passive immunotherapies utilizing monoclonal antibodies for targeting amyloid-beta (Aβ) have demonstrated promise with two recent FDA approvals. However, the pharmacological basis that differentiates clinically effective therapies from ineffective ones remains elusive. Our study offers a quantitative systems pharmacology perspective, emphasizing the significance of selectively targeting specific Aβ species and importance of antibody effector functions. This perspective sheds light on the development of more effective therapies for this devastating disease.
Collapse
Affiliation(s)
- Milica Marković
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy (M.M., Y.C.) and Lineberger Comprehensive Cancer Center, School of Medicine (Y.C.), University of North Carolina at Chapel Hill, North Carolina; Department of Biochemistry (J.M.), University of Belgrade, Faculty of Chemistry, Belgrade, Serbia; and Clinical Pharmacology and Pharmacometrics, Janssen Research & Development (W.W.), LLC, Spring House, Pennsylvania
| | - Jelica Milošević
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy (M.M., Y.C.) and Lineberger Comprehensive Cancer Center, School of Medicine (Y.C.), University of North Carolina at Chapel Hill, North Carolina; Department of Biochemistry (J.M.), University of Belgrade, Faculty of Chemistry, Belgrade, Serbia; and Clinical Pharmacology and Pharmacometrics, Janssen Research & Development (W.W.), LLC, Spring House, Pennsylvania
| | - Weirong Wang
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy (M.M., Y.C.) and Lineberger Comprehensive Cancer Center, School of Medicine (Y.C.), University of North Carolina at Chapel Hill, North Carolina; Department of Biochemistry (J.M.), University of Belgrade, Faculty of Chemistry, Belgrade, Serbia; and Clinical Pharmacology and Pharmacometrics, Janssen Research & Development (W.W.), LLC, Spring House, Pennsylvania
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy (M.M., Y.C.) and Lineberger Comprehensive Cancer Center, School of Medicine (Y.C.), University of North Carolina at Chapel Hill, North Carolina; Department of Biochemistry (J.M.), University of Belgrade, Faculty of Chemistry, Belgrade, Serbia; and Clinical Pharmacology and Pharmacometrics, Janssen Research & Development (W.W.), LLC, Spring House, Pennsylvania
| |
Collapse
|
31
|
Zhang X, Haeri M, Swerdlow RH, Wang N. Loss of Adaptive DNA Breaks in Alzheimer's Disease Brains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.566423. [PMID: 38168316 PMCID: PMC10760021 DOI: 10.1101/2023.12.11.566423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Background DNA breaks accumulate in Alzheimer's disease (AD) brains. While their role as true genomic lesions is recognized, DNA breaks also support cognitive function by facilitating the expression of activity-dependent immediate early genes (IEGs). This process involves TOP2B, a DNA topoisomerase that catalyzes the formation of DNA double-strand breaks (DSBs). Objective To characterize how AD impacts adaptive DNA breaks at nervous system genes. Methods We leveraged the ability of DNA single- and double-strand breaks to activate poly(ADP-ribose) polymerases (PARPs) that conjugate poly(ADP-ribose) (PAR) to adjacent proteins. To characterize the genomic sites harboring DNA breaks in AD brains, nuclei extracted from 3 AD and 3 non-demented (ND) autopsy brains (frontal cortex, all male donors, age 78 to 91 years of age) were analyzed through CUT&RUN in which we targeted PAR with subsequent DNA sequencing. Results Although the AD brains contained 19.9 times more PAR peaks than the ND brains, PAR peaks at nervous system genes were profoundly lost in AD brains, and the expression of these genes was downregulated. This result is consistent with our previous CUT&RUN targeting γH2AX, which marks DNA double-strand breaks (DSBs). In addition, TOP2B expression was significantly decreased in the AD brains. Conclusion Although AD brains contain a net increase in DNA breaks, adaptive DNA breaks at nervous system genes are lost in AD brains. This could potentially reflect diminished TOP2B expression and contribute to impaired neuron function and cognition in AD patients.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Institute of Reproduction and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mohammad Haeri
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Russell H. Swerdlow
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ning Wang
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Institute of Reproduction and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
32
|
Zilinskaite N, Shukla RP, Baradoke A. Use of 3D Printing Techniques to Fabricate Implantable Microelectrodes for Electrochemical Detection of Biomarkers in the Early Diagnosis of Cardiovascular and Neurodegenerative Diseases. ACS MEASUREMENT SCIENCE AU 2023; 3:315-336. [PMID: 37868357 PMCID: PMC10588936 DOI: 10.1021/acsmeasuresciau.3c00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 10/24/2023]
Abstract
This Review provides a comprehensive overview of 3D printing techniques to fabricate implantable microelectrodes for the electrochemical detection of biomarkers in the early diagnosis of cardiovascular and neurodegenerative diseases. Early diagnosis of these diseases is crucial to improving patient outcomes and reducing healthcare systems' burden. Biomarkers serve as measurable indicators of these diseases, and implantable microelectrodes offer a promising tool for their electrochemical detection. Here, we discuss various 3D printing techniques, including stereolithography (SLA), digital light processing (DLP), fused deposition modeling (FDM), selective laser sintering (SLS), and two-photon polymerization (2PP), highlighting their advantages and limitations in microelectrode fabrication. We also explore the materials used in constructing implantable microelectrodes, emphasizing their biocompatibility and biodegradation properties. The principles of electrochemical detection and the types of sensors utilized are examined, with a focus on their applications in detecting biomarkers for cardiovascular and neurodegenerative diseases. Finally, we address the current challenges and future perspectives in the field of 3D-printed implantable microelectrodes, emphasizing their potential for improving early diagnosis and personalized treatment strategies.
Collapse
Affiliation(s)
- Nemira Zilinskaite
- Wellcome/Cancer
Research UK Gurdon Institute, Henry Wellcome Building of Cancer and
Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K.
- Faculty
of Medicine, University of Vilnius, M. K. Čiurlionio g. 21, LT-03101 Vilnius, Lithuania
| | - Rajendra P. Shukla
- BIOS
Lab-on-a-Chip Group, MESA+ Institute for Nanotechnology, Max Planck
Center for Complex Fluid Dynamics, University
of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Ausra Baradoke
- Wellcome/Cancer
Research UK Gurdon Institute, Henry Wellcome Building of Cancer and
Developmental Biology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, U.K.
- Faculty
of Medicine, University of Vilnius, M. K. Čiurlionio g. 21, LT-03101 Vilnius, Lithuania
- BIOS
Lab-on-a-Chip Group, MESA+ Institute for Nanotechnology, Max Planck
Center for Complex Fluid Dynamics, University
of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
- Center for
Physical Sciences and Technology, Savanoriu 231, LT-02300 Vilnius, Lithuania
| |
Collapse
|
33
|
Taha HB, Chawla E, Bitan G. IM-MS and ECD-MS/MS Provide Insight into Modulation of Amyloid Proteins Self-Assembly by Peptides and Small Molecules. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:2066-2086. [PMID: 37607351 DOI: 10.1021/jasms.3c00065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Neurodegenerative proteinopathies are characterized by formation and deposition of misfolded, aggregated proteins in the nervous system leading to neuronal dysfunction and death. It is widely believed that metastable oligomers of the offending proteins, preceding the fibrillar aggregates found in the tissue, are the proximal neurotoxins. There are currently almost no disease-modifying therapies for these diseases despite an active pipeline of preclinical development and clinical trials for over two decades, largely because studying the metastable oligomers and their interaction with potential therapeutics is notoriously difficult. Mass spectrometry (MS) is a powerful analytical tool for structural investigation of proteins, including protein-protein and protein-ligand interactions. Specific MS tools have been useful in determining the composition and conformation of abnormal protein oligomers involved in proteinopathies and the way they interact with drug candidates. Here, we analyze critically the utilization of ion-mobility spectroscopy-MS (IM-MS) and electron-capture dissociation (ECD) MS/MS for analyzing the oligomerization and conformation of multiple amyloidogenic proteins. We also discuss IM-MS investigation of their interaction with two classes of compounds developed by our group over the last two decades: C-terminal fragments derived from the 42-residue form of amyloid β-protein (Aβ42) and molecular tweezers. Finally, we review the utilization of ECD-MS/MS for elucidating the binding sites of the ligands on multiple proteins. These approaches are readily applicable to future studies addressing similar questions and hold promise for facilitating the development of successful disease-modifying drugs against neurodegenerative proteinopathies.
Collapse
Affiliation(s)
- Hash Brown Taha
- Department of Neurology, University of California Los Angeles, California 90095, United States
- Department of Integrative Biology & Physiology, University of California Los Angeles, California 90095, United States
| | - Esha Chawla
- Department of Neurology, University of California Los Angeles, California 90095, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, California 90095, United States
| | - Gal Bitan
- Department of Neurology, University of California Los Angeles, California 90095, United States
- Brain Research Institute, University of California Los Angeles, California 90095, United States
- Molecular Biology Institute, University of California Los Angeles, California 90095, United States
| |
Collapse
|
34
|
Chaoul V, Dib EY, Bedran J, Khoury C, Shmoury O, Harb F, Soueid J. Assessing Drug Administration Techniques in Zebrafish Models of Neurological Disease. Int J Mol Sci 2023; 24:14898. [PMID: 37834345 PMCID: PMC10573323 DOI: 10.3390/ijms241914898] [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: 07/24/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 10/15/2023] Open
Abstract
Neurological diseases, including neurodegenerative and neurodevelopmental disorders, affect nearly one in six of the world's population. The burden of the resulting deaths and disability is set to rise during the next few decades as a consequence of an aging population. To address this, zebrafish have become increasingly prominent as a model for studying human neurological diseases and exploring potential therapies. Zebrafish offer numerous benefits, such as genetic homology and brain similarities, complementing traditional mammalian models and serving as a valuable tool for genetic screening and drug discovery. In this comprehensive review, we highlight various drug delivery techniques and systems employed for therapeutic interventions of neurological diseases in zebrafish, and evaluate their suitability. We also discuss the challenges encountered during this process and present potential advancements in innovative techniques.
Collapse
Affiliation(s)
- Victoria Chaoul
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (V.C.); (J.B.); (O.S.)
| | - Emanuel-Youssef Dib
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat P.O. Box 100, Lebanon; (E.-Y.D.); (C.K.)
| | - Joe Bedran
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (V.C.); (J.B.); (O.S.)
| | - Chakib Khoury
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat P.O. Box 100, Lebanon; (E.-Y.D.); (C.K.)
| | - Omar Shmoury
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (V.C.); (J.B.); (O.S.)
| | - Frédéric Harb
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat P.O. Box 100, Lebanon; (E.-Y.D.); (C.K.)
| | - Jihane Soueid
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (V.C.); (J.B.); (O.S.)
| |
Collapse
|
35
|
Unsal P, Guner M, Ozsurekci C, Balli N, Bas AO, Ozturk Y, Dikmeer A, Burkuk S, Koca M, Balci C, Dogu BB, Cankurtaran M, Halil M. Prevalence of nutrition disorders and nutrition-related conditions in older patients with Alzheimer's disease. Nutr Clin Pract 2023; 38:1142-1153. [PMID: 37076942 DOI: 10.1002/ncp.10995] [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: 12/06/2022] [Revised: 02/26/2023] [Accepted: 03/19/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUNDS Alzheimer's disease is frequently encountered with nutrition-related conditions such as malnutrition, sarcopenia, frailty, overnutrition, and micronutrient abnormalities in older patients. In this study, we aimed to evaluate the prevalence of nutrition disorders and nutrition-related conditions in the same patient group. METHODS A total of 253 older patients with Alzheimer's disease underwent comprehensive geriatric assessment, which included nutrition-related disorders, malnutrition via the Mini Nutritional Assessment-Short Form (MNA-SF), frailty via the Clinical Frailty Scale (CFS), and sarcopenia was diagnosed according to European Working Group on Sarcopenia in Older People-2 criteria. RESULTS The patients' mean age was 79.8 ± 6.5 years, and 58.1% were women. In our patients, 64.8% had malnutrition or were at risk of malnutrition; 38.3% had sarcopenia; 19.8% were prefrail; and 80.2% were frail. Malnutrition, frailty, and sarcopenia prevalence increased as the Alzheimer's disease stage progressed. Malnutrition was found to be significantly related with frailty scores via CFS (odds ratio [OR], 1.397; P = 0.0049) and muscle mass via fat-free mass index (FFMI) (OR, 0.793; P = 0.001). In logistic regression analysis, age, MNA-SF, and CFS were included in the model to detect the independent correlates of probable and confirmed sarcopenia. CFS was independently associated with probable and confirmed sarcopenia (OR, 1.822; P = 0.013; OR, 2.671; P = 0.001, respectively). Frailty was similarly related with FFMI (OR, 0.836; P = 0.031). Obesity was independently related with FFMI (OR, 0.688; P < 0.001). CONCLUSION In conclusion, nutrition disorders and nutrition-related conditions can present concurrently in patients with all stages of Alzheimer's disease; therefore, these frequent problems should be screened and diagnosed accordingly.
Collapse
Affiliation(s)
- Pelin Unsal
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Merve Guner
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Cemile Ozsurekci
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Nisa Balli
- Department of Clinical Pharmacy, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | - Arzu Okyar Bas
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Yelda Ozturk
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ayse Dikmeer
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Suna Burkuk
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Meltem Koca
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Cafer Balci
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Burcu Balam Dogu
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Mustafa Cankurtaran
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Meltem Halil
- Department of Internal Medicine, Division of Geriatric Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
36
|
Wen WC, Lin YH, Duh TH, Chen CH, Feng CH, Chen YL. Fluorescence detection of apolipoprotein E gene polymorphisms based on oligonucleotide ligation and magnetic separation. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:4710-4717. [PMID: 37680175 DOI: 10.1039/d3ay01245j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Alzheimer's disease is a progressive neurodegenerative condition that causes brain cell death and is the leading cause of dementia. Most patients with Alzheimer's disease are diagnosed with late-onset Alzheimer's disease (LOAD), with apolipoprotein E (APOE) genotypes being highly associated with the frequency of LOAD risk. A fluorescence detection system coupled with oligonucleotide ligation and magnetic separation was developed to identify two single-nucleotide polymorphisms (SNPs) for the APOE gene and recognize APOE alleles for LOAD. The system utilized a fluorescence probe with one base-discriminating nucleoside for SNP (F probe) and a perfectly complementary biotin-modified sequence against the target DNA (P probe). When the F and P probes matched the target DNA sequences, DNA ligation occurred, and ligation products were produced. Streptavidin magnetic beads were subsequently employed to remove the ligation products, and a decrease in fluorescence intensity was observed in the supernatant compared to when there was no target DNA. This system detected two SNPs of APOE alleles, namely rs429358 and rs7412. The results indicated that the R-values ((F0 - F1)/F0) for rs429358 were 0.92 ± 0.002 for the T/T target, 0.47 ± 0.004 for the T/C target and 0.11 ± 0.004 for the C/C target, respectively. The R-values for rs7412 were 0.73 ± 0.009 for the C/C target, 0.42 ± 0.001 for the C/T target and 0.16 ± 0.007 for the T/T target, respectively. F0 and F1 represent the fluorescence intensity of the F probe without and with target DNA, respectively. Based on fluorescence intensity, the fluorescence detection system was able to identify the genotypes of the APOE gene accurately to evaluate the risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Wan-Chen Wen
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Yi-Hui Lin
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung 406040, Taiwan
| | - Tsai-Hui Duh
- Department of Medicinal and Applied Chemistry, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chun-Hsien Chen
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chia-Hsien Feng
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Yen-Ling Chen
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chia-Yi 621301, Taiwan.
- Center for Nano Bio-Detection, National Chung Cheng University, Chia-Yi 621301, Taiwan
| |
Collapse
|
37
|
Teng Y, Yuan Q, Wu Y, Wu S, Su J, Zhang P, Zhang Y. Research on the Chemical Constituents against Alzheimer's Disease of the Fruits of Physalis alkekengi L. var. franchetii (Mast.) Makino. Chem Biodivers 2023; 20:e202301075. [PMID: 37505462 DOI: 10.1002/cbdv.202301075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 07/29/2023]
Abstract
Physalis alkekengi L. var. franchetii (Mast.) Makino (PA) is a natural plant which is utilised as a traditional herbal medicine. It has properties that make it effective against inflammation and free radical damage. In the present study, the major constituents of four extraction parts of the fruits of PA (PAF) were investigated by combining ultra-performance liquid chromatography with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS). The mice model of Alzheimer's disease (AD) induced by aluminum chloride (AlCl3 ) combined with D-galactose (D-gal) was established to comprehend the mechanism behind PAF's anti-AD activity from both behavioural and pathological perspectives. The results showed that four extraction parts of PAF (PAFE) had favorable anti-AD effects and the ethyl acetate (EA) group showed the best activity. UPLC-Q-TOF-MS analysis identified Physalin B, Nobiletin and Caffeic acid as the main anti-AD active constituents in EA extract. This study reveals that PAF can reduce neuroinflammatory damage by inhibiting p38 mitogen-activated protein kinase (p38 MAPK) signaling pathway, which is the theoretical basis for clinical development and utilization of PAF in AD therapy.
Collapse
Affiliation(s)
- Yang Teng
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
- Heilongjiang Pharmaceutical Research Institute, Jiamusi, 154007, China
| | - Qi Yuan
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
| | - You Wu
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
| | - Shuang Wu
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
| | - Jin Su
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
- Heilongjiang Pharmaceutical Research Institute, Jiamusi, 154007, China
| | - Pengxia Zhang
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
- Heilongjiang Pharmaceutical Research Institute, Jiamusi, 154007, China
| | - Yu Zhang
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, China
- Heilongjiang Pharmaceutical Research Institute, Jiamusi, 154007, China
| |
Collapse
|
38
|
Tavasoli A, Gelman BB, Marra CM, Clifford DB, Iudicello JE, Rubin LH, Letendre SL, Tang B, Ellis RJ. Increasing Neuroinflammation Relates to Increasing Neurodegeneration in People with HIV. Viruses 2023; 15:1835. [PMID: 37766242 PMCID: PMC10536802 DOI: 10.3390/v15091835] [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: 08/08/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND HIV infection causes neuroinflammation and immune activation (NIIA) and systemic inflammation and immune activation (SIIA), which in turn drive neurodegeneration (ND). Cross-sectionally, higher levels of NIIA biomarkers correlate with increased biomarkers of ND. A more convincing confirmation would be a longitudinal demonstration. METHODS PWH in the US multisite CHARTER Aging project were assessed at a baseline visit and after 12 years using standardized evaluations. We measured a panel of 14 biomarkers of NIIA, SIIA, and ND in plasma and CSF at two time points and calculated changes from baseline to the 12-year visit. Factor analysis yielded simplified indices of NIIA, SIIA, and ND. RESULTS The CSF NIIA factor analysis yielded Factor1 loading on soluble tumor necrosis factor type-2 (sTNFR-II) and neopterin, and Factor2, loading on MCP1, soluble CD14, and IL-6. The SIIA factor analysis yielded Factor1 loading on CRP, D-dimer, and Neopterin; Factor2 loading on sTNFR-II. The ND analysis yielded Factor1 loading on Phosphorylated tau (p-tau) and Aβ42; Factor2 loading on NFL. NIIA Factor1, but not Factor2, correlated with increases in CSF NFL (r = 0.370, p = 0.0002). CONCLUSIONS Increases in NIIA and SIIA in PWH were associated with corresponding increases in ND, suggesting that reducing neuro/systemic inflammation might slow or reverse neurodegeneration.
Collapse
Affiliation(s)
- Azin Tavasoli
- Department of Neurosciences, University of California San Diego, San Diego, CA 92093, USA;
| | - Benjamin B. Gelman
- Department of Pathology, University of Texas at Galveston, Galveston, TX 77555, USA;
| | - Christina M. Marra
- Department of Medicine, University of Washington, Seattle, WA 98195, USA;
| | - David B. Clifford
- Department of Neurology, Washington University in St. Louis, St. Louis, MO 63130, USA;
| | - Jennifer E. Iudicello
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA; (J.E.I.); (B.T.)
| | - Leah H. Rubin
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Scott L. Letendre
- Department of Medicine, University of California San Diego, San Diego, CA 92093, USA;
| | - Bin Tang
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA; (J.E.I.); (B.T.)
| | - Ronald J. Ellis
- Department of Neurosciences, University of California San Diego, San Diego, CA 92093, USA;
| |
Collapse
|
39
|
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
|
40
|
Feter N, de Paula D, Leite JS, Caputo EL, Rombaldi AJ. The association of aerobic and muscular fitness with cognitive impairment: Findings from a nationally representative survey. Psychiatry Res 2023; 326:115360. [PMID: 37494879 DOI: 10.1016/j.psychres.2023.115360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 07/28/2023]
Abstract
This study aimed to characterize the combined association between cardiorespiratory fitness (CRF), muscular strength, and cognitive outcomes in middle-aged and older adults from low and middle-income countries (LMICs). We analyzed cross-sectional, population-based data from adults aged 50 years or older from six LMICs. Mild cognitive impairment (MCI) was defined according to the National Institute on Aging-Alzheimer's Association criteria. Estimated CRF (eCRF) was calculated using previously validated, sex-specific equations. Handgrip strength (HS) was used as an indicator of muscular strength. We used linear and robust Poisson regression models to examine the associations between eCRF, HS, and MCI. Data from 28,339 adults (63.1 [9.5] years) were analyzed. Participants with low eCRF (PR: 1.45; 95%CI: 1.11, 1.90) and HS (PR: 1.92; 95%CI: 1.79, 2.04) were more prone to have MCI. Participants with low HS showed higher likelihood of MCI than those with preserved HS through the CRF range; however, this difference was not seen among highly fit individuals (10 METs or higher). Each 1-MET (PR: 0.77; 95%CI: 0.67, 0.86) and 5-kgf (PR: 0.63; 95%CI: 0.48, 0.79) increase was associated with a reduction in the likelihood of MCI. eCRF and HS were strongly and independently associated with MCI in middle-aged and older adults.
Collapse
Affiliation(s)
- Natan Feter
- Post Graduate Program in Epidemiology, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Danilo de Paula
- Post Graduate Program in Epidemiology, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jayne S Leite
- Post Graduate Program in Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo L Caputo
- Center for Evidence Synthesis in Health, School of Public Health, Brown University, Providence, Rhode Island, United States
| | - Airton J Rombaldi
- Post Graduate Program in Physical Education, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| |
Collapse
|
41
|
Thomas S, Prendergast GC. Gut-brain connections in neurodegenerative disease: immunotherapeutic targeting of Bin1 in inflammatory bowel disease and Alzheimer's disease. Front Pharmacol 2023; 14:1183932. [PMID: 37521457 PMCID: PMC10372349 DOI: 10.3389/fphar.2023.1183932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/13/2023] [Indexed: 08/01/2023] Open
Abstract
Longer lifespan produces risks of age-associated neurodegenerative disorders such as Alzheimer's disease (AD), which is characterized by declines in memory and cognitive function. The pathogenic causes of AD are thought to reflect a progressive aggregation in the brain of amyloid plaques composed of beta-amyloid (Aß) peptides and neurofibrillary tangles composed of phosphorylated tau protein. Recently, long-standing investigations of the Aß disease hypothesis gained support via a passive immunotherapy targeting soluble Aß protein. Tau-targeting approaches using antibodies are also being pursued as a therapeutic approach to AD. In genome-wide association studies, the disease modifier gene Bin1 has been identified as a top risk factor for late-onset AD in human populations, with recent studies suggesting that Bin1 binds tau and influences its extracellular deposition. Interestingly, before AD emerges in the brain, tau levels rise in the colon, where Bin1-a modifier of tissue barrier function and inflammation-acts to promote inflammatory bowel disease (IBD). This connection is provocative given clinical evidence of gut-brain communication in age-associated neurodegenerative disorders, including AD. In this review, we discuss a Bin1-targeting passive immunotherapy developed in our laboratory to treat IBD that may offer a strategy to indirectly reduce tau deposition and limit AD onset or progression.
Collapse
|
42
|
Nguyen TT, Nguyen-Thi PT, Nguyen THA, Ho TT, Tran NMA, Van Vo T, Van Vo G. Recent Advancements in Nanomaterials: A Promising Way to Manage Neurodegenerative Disorders. Mol Diagn Ther 2023; 27:457-473. [PMID: 37217723 DOI: 10.1007/s40291-023-00654-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2023] [Indexed: 05/24/2023]
Abstract
Neurodegenerative diseases (NDs) such as dementia, Alzheimer's disease, Parkinson's disease, frontotemporal dementia, and amyotrophic lateral sclerosis are some of the most prevalent disorders currently afflicting healthcare systems. Many of these diseases share similar pathological hallmarks, including elevated oxidative stress, mitochondrial dysfunction, protein misfolding, excitotoxicity, and neuroinflammation, all of which contribute to the deterioration of the nervous system's structure and function. The development of diagnostic and therapeutic materials in the monitoring and treatment of these diseases remains challenging. One of the biggest challenges facing therapeutic and diagnostic materials is the blood-brain barrier (BBB). The BBB is a multifunctional membrane possessing a plethora of biochemical, cellular, and immunological features that ensure brain homeostasis by preventing the entry and accumulation of unwanted compounds. With regards to neurodegenerative diseases, the recent application of tailored nanomaterials (nanocarriers and nanoparticles) has led to advances in diagnostics and therapeutics. In this review, we provide an overview of commonly used nanoparticles and their applications in NDs, which may offer new therapeutic strategies for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City, 71420, Vietnam
| | | | - Thi Hong Anh Nguyen
- Ho Chi Minh City University of Food Industry (HUFI), 140 Le Trong Tan Street, Tay Thanh Ward, Tan Phu District, Ho Chi Minh City, 700000, Vietnam
| | - Thanh-Tam Ho
- Institute for Global Health Innovations, Duy Tan University, Da Nang, 550000, Vietnam.
- Faculty of Pharmacy, Duy Tan University, Da Nang, 550000, Vietnam.
| | - Nguyen-Minh-An Tran
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City, 71420, Vietnam
| | - Toi Van Vo
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, 700000, Vietnam.
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
| | - Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
- Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University, Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
43
|
Kim S, Sharma C, Jung UJ, Kim SR. Pathophysiological Role of Microglial Activation Induced by Blood-Borne Proteins in Alzheimer's Disease. Biomedicines 2023; 11:biomedicines11051383. [PMID: 37239054 DOI: 10.3390/biomedicines11051383] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
The blood-brain barrier (BBB) restricts entry of neurotoxic plasma components, blood cells, and pathogens into the brain, leading to proper neuronal functioning. BBB impairment leads to blood-borne protein infiltration such as prothrombin, thrombin, prothrombin kringle-2, fibrinogen, fibrin, and other harmful substances. Thus, microglial activation and release of pro-inflammatory mediators commence, resulting in neuronal damage and leading to impaired cognition via neuroinflammatory responses, which are important features observed in the brain of Alzheimer's disease (AD) patients. Moreover, these blood-borne proteins cluster with the amyloid beta plaque in the brain, exacerbating microglial activation, neuroinflammation, tau phosphorylation, and oxidative stress. These mechanisms work in concert and reinforce each other, contributing to the typical pathological changes in AD in the brain. Therefore, the identification of blood-borne proteins and the mechanisms involved in microglial activation and neuroinflammatory damage can be a promising therapeutic strategy for AD prevention. In this article, we review the current knowledge regarding the mechanisms of microglial activation-mediated neuroinflammation caused by the influx of blood-borne proteins into the brain via BBB disruption. Subsequently, the mechanisms of drugs that inhibit blood-borne proteins, as a potential therapeutic approach for AD, along with the limitations and potential challenges of these approaches, are also summarized.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Chanchal Sharma
- School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Sang Ryong Kim
- School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
44
|
Liu M, Zhu L, Guo YJ, Zhang SS, Jiang L, Zhang Y, Chao FL, Tang Y. The effects of voluntary running exercise on the astrocytes of the medial prefrontal cortex in APP/PS1 mice. J Comp Neurol 2023. [PMID: 37146123 DOI: 10.1002/cne.25485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 05/07/2023]
Abstract
Pathological changes in the medial prefrontal cortex (mPFC) and astrocytes are closely associated with Alzheimer's disease (AD). Voluntary running has been found to effectively delay AD. However, the effects of voluntary running on mPFC astrocytes in AD are unclear. A total of 40 10-month-old male amyloid precursor protein/presenilin 1 (APP/PS1) mice and 40 wild-type (WT) mice were randomly divided into control and running groups, and the running groups underwent voluntary running for 3 months. Mouse cognition was assessed by the novel object recognition (NOR), Morris water maze (MWM), and Y maze tests. The effects of voluntary running on mPFC astrocytes were investigated using immunohistochemistry, immunofluorescence, western blotting, and stereology. APP/PS1 mice performed significantly worse than WT mice in the NOR, MWM, and Y maze tests, and voluntary running improved the performance of APP/PS1 mice in these tests. The total number of mPFC astrocytes was increased, cell bodies were enlarged, and protrusion number and length were increased in AD mice compared with WT mice, but there was no difference in component 3 (C3) levels in the mPFC (total mPFC level); however, C3 and S100B levels in astrocytes were increased in AD mice. Voluntary running reduced the total number of astrocytes and S100B levels in astrocytes and increased the density of PSD95+ puncta in direct contact with astrocyte protrusions in the APP/PS1 mouse mPFC. Three months of voluntary running inhibited astrocyte hyperplasia and S100B expression in astrocytes, increased the density of synapses in contact with astrocytes, and improved cognitive function in APP/PS1 mice.
Collapse
Affiliation(s)
- Mei Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Lin Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Yi-Jing Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Shan-Shan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Army Medical University, Chongqing, P. R. China
| | - Lin Jiang
- Laboratory Teaching & Management Center, Chongqing Medical University, Chongqing, P. R. China
| | - Yi Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Feng-Lei Chao
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
45
|
Varte V, Munkelwitz JW, Rincon-Limas DE. Insights from Drosophila on Aβ- and tau-induced mitochondrial dysfunction: mechanisms and tools. Front Neurosci 2023; 17:1184080. [PMID: 37139514 PMCID: PMC10150963 DOI: 10.3389/fnins.2023.1184080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia in older adults worldwide. Sadly, there are no disease-modifying therapies available for treatment due to the multifactorial complexity of the disease. AD is pathologically characterized by extracellular deposition of amyloid beta (Aβ) and intracellular neurofibrillary tangles composed of hyperphosphorylated tau. Increasing evidence suggest that Aβ also accumulates intracellularly, which may contribute to the pathological mitochondrial dysfunction observed in AD. According with the mitochondrial cascade hypothesis, mitochondrial dysfunction precedes clinical decline and thus targeting mitochondria may result in new therapeutic strategies. Unfortunately, the precise mechanisms connecting mitochondrial dysfunction with AD are largely unknown. In this review, we will discuss how the fruit fly Drosophila melanogaster is contributing to answer mechanistic questions in the field, from mitochondrial oxidative stress and calcium dysregulation to mitophagy and mitochondrial fusion and fission. In particular, we will highlight specific mitochondrial insults caused by Aβ and tau in transgenic flies and will also discuss a variety of genetic tools and sensors available to study mitochondrial biology in this flexible organism. Areas of opportunity and future directions will be also considered.
Collapse
Affiliation(s)
- Vanlalrinchhani Varte
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
| | - Jeremy W. Munkelwitz
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
| | - Diego E. Rincon-Limas
- Department of Neurology, McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Genetics Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
46
|
Imai A, Matsuoka T, Narumoto J. Emotional Dysregulation in Mild Behavioral Impairment Is Associated with Reduced Cortical Thickness in the Right Supramarginal Gyrus. J Alzheimers Dis 2023; 93:521-532. [PMID: 37038811 DOI: 10.3233/jad-220948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
BACKGROUND Mild behavioral impairment (MBI) has attracted attention as a possible precursor symptom of dementia, but its neural basis has not been fully investigated. OBJECTIVE We aimed to investigate the relationship between MBI and surface area, cortical thickness, and volume in the temporal and parietal lobes, which are strongly associated with dementia and emotional disorders. METHODS This retrospective study evaluated 123 participants: 90 with mild cognitive impairment (MCI), 13 with subjective cognitive decline (SCD), and 20 cognitively healthy (CH). Using analysis of covariance (ANCOVA) with sex, age, and MMSE score as covariates, cortical thickness, surface area, and volume in 10 regions were compared between groups with and without MBI. Groups with MBI emotional dysregulation were also compared with groups without MBI. RESULTS ANCOVA revealed significantly smaller cortical thickness in the MBI group's right parahippocampal (p = 0.01) and supramarginal gyri (p = 0.002). After multiple comparison correction, only the right supramarginal gyrus was significantly smaller (p = 0.02). When considering only MBI emotional dysregulation, the right parahippocampal and supramarginal gyrus' cortical thicknesses were significantly smaller in this MBI group (p = 0.03, 0.01). However, multiple comparison correction identified no significant differences (p = 0.14, 0.11). CONCLUSION Overall MBI and the emotional dysregulation domains were associated with reduced cortical thickness in the right parahippocampal and supramarginal gyri. Since neurodegeneration in the medial temporal and parietal lobe precedes early Alzheimer's disease (AD), MBI, particularly emotion dysregulation, may predict early AD below the diagnostic threshold.
Collapse
Affiliation(s)
- Ayu Imai
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Teruyuki Matsuoka
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jin Narumoto
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
47
|
Eisenmenger LB, Peret A, Famakin BM, Spahic A, Roberts GS, Bockholt JH, Johnson KM, Paulsen JS. Vascular contributions to Alzheimer's disease. Transl Res 2023; 254:41-53. [PMID: 36529160 PMCID: PMC10481451 DOI: 10.1016/j.trsl.2022.12.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and is characterized by progressive neurodegeneration and cognitive decline. Understanding the pathophysiology underlying AD is paramount for the management of individuals at risk of and suffering from AD. The vascular hypothesis stipulates a relationship between cardiovascular disease and AD-related changes although the nature of this relationship remains unknown. In this review, we discuss several potential pathological pathways of vascular involvement in AD that have been described including dysregulation of neurovascular coupling, disruption of the blood brain barrier, and reduced clearance of metabolite waste such as beta-amyloid, a toxic peptide considered the hallmark of AD. We will also discuss the two-hit hypothesis which proposes a 2-step positive feedback loop in which microvascular insults precede the accumulation of Aß and are thought to be at the origin of the disease development. At neuroimaging, signs of vascular dysfunction such as chronic cerebral hypoperfusion have been demonstrated, appearing early in AD, even before cognitive decline and alteration of traditional biomarkers. Cerebral small vessel disease such as cerebral amyloid angiopathy, characterized by the aggregation of Aß in the vessel wall, is highly prevalent in vascular dementia and AD patients. Current data is unclear whether cardiovascular disease causes, precipitates, amplifies, precedes, or simply coincides with AD. Targeted imaging tools to quantitatively evaluate the intracranial vasculature and longitudinal studies in individuals at risk for or in the early stages of the AD continuum could be critical in disentangling this complex relationship between vascular disease and AD.
Collapse
Affiliation(s)
- Laura B Eisenmenger
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Anthony Peret
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Bolanle M Famakin
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Alma Spahic
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Grant S Roberts
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jeremy H Bockholt
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, Georgia
| | - Kevin M Johnson
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jane S Paulsen
- Department of Neurology, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
48
|
Mei Y, Li Y, Cheng Y, Gao L. The effect of gastric bypass surgery on cognitive function of Alzheimer's disease and the role of GLP1-SGLT1 pathway. Exp Neurol 2023; 363:114377. [PMID: 36893833 DOI: 10.1016/j.expneurol.2023.114377] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/20/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVE Gastric bypass surgery has been shown to improve metabolic profiles via GLP1, which may also have cognitive benefits for Alzheimer's disease (AD) patients. However, the exact mechanism requires further investigation. METHODS Roux-en-Y gastric bypass or sham surgery was performed on APP/PS1/Tau triple transgenic mice (an AD mice model) or wild type C57BL/6 mice. Morris Water Maze (MWM) test was used to evaluate the cognitive function of mice and animal tissue samples were obtained for measurements two months after the surgery. Additionally, STC-1 intestine cells were treated with siTAS1R2 and siSGLT1, and HT22 nerve cells were treated with Aβ, siGLP1R, GLP1 and siSGLT1 in vitro to explore the role of GLP1-SGLT1 related signaling pathway in cognitive function. RESULTS The MWM test showed that bypass surgery significantly improved cognitive function in AD mice as measured by navigation and spatial probe tests. Moreover, bypass surgery reversed neurodegeneration, down-regulated hyperphosphorylation of Tau protein and Aβ deposition, improved glucose metabolism, and up-regulated the expression of GLP1, SGLT1, and TAS1R2/3 in the hippocampus. Furthermore, GLP1R silencing down-regulated SGLT1 expression, whereas SGLT1 silencing increased Tau protein deposition and exacerbated dysregulated of glucose metabolism in HT22 cells. However, RYGB did not alter the level of GLP1 secretion in the brainstem (where central GLP1 is mainly produced). Additionally, GLP1 expression was upregulated by RYGB via TAS1R2/3-SGLT1 activation sequentially in the small intestine. CONCLUSION RYGB surgery could improve cognition function in AD mice through facilitating glucose metabolism and reducing Tau phosphorylation and Aβ deposition in the hippocampus, mediated by peripheral serum GLP1 activation of SGLT1 in the brain. Furthermore, RYGB increased GLP1 expression through sequential activation of TAS1R2/TAS1R3 and SGLT1 in the small intestine.
Collapse
Affiliation(s)
- Yingna Mei
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, China
| | - Yubing Li
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, China
| | - Yanxiang Cheng
- Department of Obstetrics & Gynecology, Renmin Hospital of Wuhan University, China.
| | - Ling Gao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, China.
| |
Collapse
|
49
|
In Vitro Antiviral Effect and Potential Neuroprotection of Salvadora persica L. Stem Bark Extract against Lipopolysaccharides-Induced Neuroinflammation in Mice: LC-ESI-MS/MS Analysis of the Methanol Extract. Pharmaceuticals (Basel) 2023; 16:ph16030398. [PMID: 36986497 PMCID: PMC10058283 DOI: 10.3390/ph16030398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroinflammation is a serious immunomodulatory complex disorder that causes neurological and somatic ailments. The treatment of brain inflammation with new drugs derived from natural sources is a significant therapeutic goal. Utilizing LC-ESI-MS/MS analysis, the active constituents of Salvadora persica extract (SPE) were identified tentatively as exerting antioxidant and anti-inflammatory effects in natural medicine. Herein, we determined the antiviral potential of SPE against herpes simplex virus type 2 (HSV-2) using the plaque assay. HSV-2 is a neurotropic virus that can cause neurological diseases. SPE exhibited promising antiviral potential with a half-maximal cytotoxic concentration (CC50) of 185.960 ± 0.1 µg/mL and a half-maximal inhibitory concentration (IC50) of 8.946 ± 0.02 µg/mL. The in vivo study of the SPE impact against lipopolysaccharide (LPS)-induced neuroinflammation was performed using 42 mice divided into seven groups. All groups were administered LPS (0.25 mg/kg) intraperitoneally, except for the normal and SPE groups 1 and 2. Groups 5, 6, and 7 received 100, 200, and 300 mg/kg SPE. It was revealed that SPE inhibited acetylcholinesterase in the brain. It increased superoxide dismutase and catalase while decreasing malondialdehyde, which explains its antioxidative stress activity. SPE downregulated the gene expression of the inducible nitric oxide synthase, as well as the apoptotic markers (caspase-3 and c-Jun). In addition, it decreased the expression of the proinflammatory cytokines (interleukin-6 and tumor necrosis factor-alpha). Mice administered SPE (300 mg/kg) with LPS exhibited normal neurons in the cerebral cortices, hippocampus pyramidal layer, and cerebellum, as determined by the histopathological analysis. Therefore, using S. persica to prevent and treat neurodegeneration could be a promising new therapeutic strategy to be explored.
Collapse
|
50
|
Canet G, Zussy C, Hernandez C, Maurice T, Desrumaux C, Givalois L. The pathomimetic oAβ25–35 model of Alzheimer's disease: Potential for screening of new therapeutic agents. Pharmacol Ther 2023; 245:108398. [PMID: 37001735 DOI: 10.1016/j.pharmthera.2023.108398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly, currently affecting more than 40 million people worldwide. The two main histopathological hallmarks of AD were identified in the 1980s: senile plaques (composed of aggregated amyloid-β (Aβ) peptides) and neurofibrillary tangles (composed of hyperphosphorylated tau protein). In the human brain, both Aβ and tau show aggregation into soluble and insoluble oligomers. Soluble oligomers of Aβ include their most predominant forms - Aβ1-40 and Aβ1-42 - as well as shorter peptides such as Aβ25-35 or Aβ25-35/40. Most animal models of AD have been developed using transgenesis, based on identified human mutations. However, these familial forms of AD represent less than 1% of AD cases. In this context, the idea emerged in the 1990s to directly inject the Aβ25-35 fragment into the rodent brain to develop an acute model of AD that could mimic the disease's sporadic forms (99% of all cases). This review aims to: (1) summarize the biological activity of Aβ25-35, focusing on its impact on the main structural and functional alterations observed in AD (cognitive deficits, APP misprocessing, tau system dysfunction, neuroinflammation, oxidative stress, cholinergic and glutamatergic alterations, HPA axis dysregulation, synaptic deficits and cell death); and (2) confirm the interest of this pathomimetic model in AD research, as it has helped identify and characterize many molecules (marketed, in clinical development, and in preclinical testing), and to the development of alternative approaches for AD prevention and therapy. Today, the Aβ25-35 model appears as a first-intent choice model to rapidly screen the symptomatic or neuroprotective potencies of new compounds, chemical series, or innovative therapeutic strategies.
Collapse
|