1
|
Hu H, Wan S, Hu Y, Wang Q, Li H, Zhang N. Deciphering the role of APOE in cerebral amyloid angiopathy: from genetic insights to therapeutic horizons. Ann Med 2025; 57:2445194. [PMID: 39745195 DOI: 10.1080/07853890.2024.2445194] [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: 07/16/2024] [Revised: 10/26/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Cerebral amyloid angiopathy (CAA), characterized by the deposition of amyloid-β (Aβ) peptides in the walls of medium and small vessels of the brain and leptomeninges, is a major cause of lobar hemorrhage in elderly individuals. Among the genetic risk factors for CAA that continue to be recognized, the apolipoprotein E (APOE) gene is the most significant and prevalent, as its variants have been implicated in more than half of all patients with CAA. While the presence of the APOE ε4 allele markedly increases the risk of CAA, the ε2 allele confers a protective effect relative to the common ε3 allele. These allelic variants encode three APOE isoforms that differ at two amino acid positions. The primary physiological role of APOE is to mediate lipid transport in the brain and periphery; however, it has also been shown to be involved in a wide array of biological functions, particularly those involving Aβ, in which it plays a known role in processing, production, aggregation, and clearance. The challenges posed by the reliance on postmortem histological analyses and the current absence of an effective intervention underscore the urgency for innovative APOE-targeted strategies for diagnosing CAA. This review not only deepens our understanding of the impact of APOE on the pathogenesis of CAA but can also help guide the exploration of targeted therapies, inspiring further research into the therapeutic potential of APOE.
Collapse
Affiliation(s)
- Hantian Hu
- Tianjin Medical University, Tianjin, China
| | - Siqi Wan
- Tianjin Medical University, Tianjin, China
| | - Yuetao Hu
- Tianjin Medical University, Tianjin, China
| | - Qi Wang
- Tianjin Medical University, Tianjin, China
| | - Hanyu Li
- Tianjin Medical University, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Trompeta C, Gasca-Salas C, Rodríguez-Rojas R, Fernández-Rodríguez B, Clarimón J, García-Cañamaque L, Guida P, Hernández-Fernández F, Sánchez-Juan P, Olazarán J, Vela-Desojo L. Apolipoprotein E genotypes and amyloid burden associated with cognitive decline in non-demented Parkinson's disease patients: A preliminary analysis. J Neurol Sci 2025; 473:123510. [PMID: 40300362 DOI: 10.1016/j.jns.2025.123510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/07/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Cognitive impairment is a common non-motor manifestation in Parkinson's disease (PD). Amyloidosis seems to accelerate the onset of dementia in PD. The Apolipoprotein (APOE) ε4 genotype is linked to amyloid aggregation and has been suggested to be a candidate for development of PD dementia, while other APOE alleles have been less investigated. METHODS Forty-eight non-demented PD patients were included (ε2/ε3 carriers: n = 8; ε3/ε3 carriers: n = 35; and ε3/ε4 carriers: n = 5). Patients underwent APOE genotyping, a neuropsychological exam and a [18F]-Flutemetamol PET. Statistical Parametric Mapping 12 was used to analyze regional amyloid differences by APOE genotypes. Cognitive assessment was re-administered after follow-up. RESULTS APOE ε3/ε4 carriers showed significant amyloid load in left middle occipital areas in comparison with APOE ε3/ε3, whereas APOE ε3/ε4 exhibited several areas of amyloid accumulation in comparison with APOE ε2/ε3 that disappeared after family-wise error cluster level correction, but which hold in nonparametric analyses. No amyloid load differences were found between APOE ε2/ε3 and APOE ε3/ε3. The frequency of progression of cognitive decline was 80 % in ε3/ε4, 50 % in ε2/ε3, and 22.86 % in ε3/ε3 carriers. CONCLUSIONS This compared regional amyloid deposition and cognitive evolution among APOE genotypes. Our preliminary findings suggest an association between APOE ε4 and amyloid burden in posterior cortical areas in non-demented PD and a tendency to a higher frequency of cognitive decline. Larger studies should explore cognition and amyloid burden in APOE ε2 carriers.
Collapse
Affiliation(s)
- Clara Trompeta
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; PhD Program in Health Sciences, University of Alcala de Henares, Alcala de Henares, Spain
| | - Carmen Gasca-Salas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; University CEU-San Pablo, Madrid, Spain; Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| | - Rafael Rodríguez-Rojas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Beatriz Fernández-Rodríguez
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Department of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
| | | | - Lina García-Cañamaque
- Nuclear Medicine Department, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Pasqualina Guida
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; PhD Program in Neuroscience, Autonoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Frida Hernández-Fernández
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Department of Nursing and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid,Madrid, Spain
| | - Pascual Sánchez-Juan
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Alzheimer's Centre Reina Sofia-CIEN Foundation-ISCIII, 28031 Madrid, Spain
| | - Javier Olazarán
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | |
Collapse
|
3
|
Johnson CN, Evans MR, Blankenship AE, John CS, Rekowski MJ, Washburn MP, Phan A, Gouvion CM, Haeri M, Swerdlow RH, Geiger PC, Morris JK. Human skeletal muscle mitochondrial pathways are impacted by a neuropathologic diagnosis of Alzheimer's disease. Neurobiol Dis 2025; 210:106916. [PMID: 40250718 DOI: 10.1016/j.nbd.2025.106916] [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: 02/12/2025] [Revised: 03/21/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Alzheimer's disease (AD) is associated with reduced lean mass and impaired skeletal muscle mitochondrial and motor function. Although primary mitochondrial defects in AD may underlie these findings, molecular alterations in AD have not been thoroughly examined in human skeletal muscle. Here, we used two human skeletal muscle types, quadriceps (n = 81) and temporalis (n = 66), to compare the proteome of individuals with a neuropathologic AD diagnosis based on AD Neuropathologic Change (ADNPC+: n = 54 temporalis, 44 quadriceps) to controls (ADNPC-: n = 27 temporalis, 22 quadriceps). We determined the effects of ADNPC status within each muscle and within apolipoprotein E4 (APOE4) carriers and APOE4 non-carriers. Pathways that support mitochondrial metabolism, including oxidative phosphorylation, were downregulated in skeletal muscle of ADNPC+ versus ADNPC- individuals. Similar mitochondrial effects were observed across muscle types and APOE4 carrier groups, but nearly four times as many proteins were altered in temporalis versus quadriceps tissue and mitochondrial effects were most pronounced in APOE4 carriers compared to APOE4 non-carriers. Of all detected oxidative phosphorylation proteins, the expression of ∼29-61 % (dependent on muscle/APOE4 carrier group) significantly correlated with AD progression, ranked by Clinical Dementia Rating and ADNPC scores. Of these, 23 proteins decreased in expression with greater AD progression in all skeletal muscle type and APOE4 carrier groups. This is the first study to assess differences in the human skeletal muscle proteome in the context of AD. Our work shows that systemic mitochondrial alterations in AD extend to skeletal muscle and these effects are amplified by APOE4 and correlate with AD progression.
Collapse
Affiliation(s)
- Chelsea N Johnson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Mara R Evans
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anneka E Blankenship
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
| | - Casey S John
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Michaella J Rekowski
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Michael P Washburn
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Andy Phan
- Bruker Daltonics, Inc, Billerica, MA 01821, USA.
| | - Cynthia M Gouvion
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Mohammad Haeri
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Paige C Geiger
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Jill K Morris
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
4
|
Claus JJ, Rosbergen MT, Labrecque JA, Vernooij MW, Wolters FJ, Ikram MA. Mediation of the Association Between APOE ε4 Genotype, Cognition, and Dementia by Neuropathology Imaging Markers in the Rotterdam Study. Neurology 2025; 104:e213679. [PMID: 40344552 PMCID: PMC12063243 DOI: 10.1212/wnl.0000000000213679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/19/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Insight into APOE-related pathways is important to unravel pathophysiology and identify therapeutic targets against late-life cognitive decline. We aimed to estimate mediators of APOE ε4 on cognition and dementia through different disease markers on structural in vivo brain imaging. METHODS All participants from the population-based Rotterdam Study who underwent brain MRI between 2005 and 2009 were included. Cognition was assessed cross-sectionally during center visits, and participants were followed up for incident dementia until January 1, 2020. Imaging markers included hippocampal volume (HV), volume of white matter hyperintensities (WMHs), Alzheimer disease-specific regional cortical thickness, and presence of ≥2 cerebral microbleeds. We performed causal mediation analyses to decompose the total effect of APOE ε4 carriership on cognition and dementia into natural direct and indirect effects and corresponding percentage mediated. We adjusted models for potential confounders. RESULTS Among 5,510 participants (mean age at time of MRI scan: 65.0 [±10.9] years, 55.0% women), 349 developed dementia, of whom 148 were ε4 carriers. Carriers of ε4 had slightly lower Z-scores for global cognition (β = -0.02 [-0.07 to 0.02], age-related cognitive decline = 4.4 months), with 7% (β = -0.00 [0.00-0.00]) of this association mediated by HV and 4% (β = -0.00 [-0.01 to 0.00]) by cortical thickness. In total, an estimated 25% of the effect of ε4 on cognition was mediated by microbleeds (p value = 0.24, [β = -0.00 {-0.01 to 0.00}]) and 12% by WMHs (p value = 0.44, [β = -0.00 {-0.01 to 0.00}]). In multiple mediator analyses, WMHs and microbleeds together accounted for 27% of the mediated effect of APOE ε4 on cognition (p value = 0.48). Carriers of ε4 had higher risk of incident dementia (HR 2.35 [95% CI 2.06-2.65]). For dementia, there was little to no evidence of mediation by either HV (3%, p value = 0.09, OR = 1.01 [1.00-1.03]) or regional cortical thickness (0%, p value = 0.79, OR = 1.00 [0.99-1.02]). In total, 1% of the effect of ε4 on dementia was mediated by WMHs (p value 0.29, OR = 1.00 [1.00-1.02]) and 5% by microbleeds (p value = 0.06), OR = 1.03 (1.00-1.07). In multiple mediator analyses, all 4 imaging markers together explained 6% of the mediated effect on incident dementia (p value = 0.04). DISCUSSION In this population-based cohort study, we found that an estimated one-fourth of the effect of APOE ε4 on cognition is mediated by structural brain imaging markers, driven mainly by cerebral microbleeds. For dementia, mediation by these markers was limited.
Collapse
Affiliation(s)
- Jacqueline J Claus
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; and
- Department of Radiology & Nuclear Medicine and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Mathijs T Rosbergen
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; and
- Department of Radiology & Nuclear Medicine and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jeremy A Labrecque
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; and
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; and
- Department of Radiology & Nuclear Medicine and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Frank J Wolters
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; and
- Department of Radiology & Nuclear Medicine and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Mohammad Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; and
| |
Collapse
|
5
|
Li W, Ma S, Li M. Intervention Role of APOE in CNS Diseases: APOE Actions and APOE Neurogenesis Capability. Mol Neurobiol 2025:10.1007/s12035-025-05028-8. [PMID: 40402407 DOI: 10.1007/s12035-025-05028-8] [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: 03/23/2024] [Accepted: 05/02/2025] [Indexed: 05/23/2025]
Abstract
Neurogenesis is a biological process in which new neurons are generated from neural stem cells (NSCs) in specific neural niches in the brain. Impaired neurogenesis, characterized by the progressive loss of neurons, leads to cognitive and motor disabilities and is a hallmark of central nervous system (CNS) diseases. Conversely, enhancing neurogenesis has been shown to alleviate the symptoms of CNS diseases. Apolipoprotein E (APOE) is a protein that plays various biological roles in CNS diseases. Emerging research indicates that APOE is involved in adult neurogenesis, which is crucial for maintaining the neural progenitor pool in the dentate gyrus (DG) and synaptic activity. Therefore, APOE could be a therapeutic target for promoting neurogenesis in the treatment and intervention of CNS diseases. In this context, we present a comprehensive overview of the clinical evidence supporting the role of APOE in CNS diseases on the basis of a meta-analysis. We also discuss the neurogenic potential of APOE, which has significant implications not only for understanding the biological underpinnings of neurological diseases but also for developing novel treatment strategies for CNS diseases.
Collapse
Affiliation(s)
- Wenhua Li
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacologyof, Traditional Chinese Medicine Formulae, Institute of Traditional Chinese Medicine , Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Suya Ma
- Guang'anmen Hospital, China, Academy of Chinese Medicine Sciences , Beijing, 100053, China
| | - Min Li
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| |
Collapse
|
6
|
Miyashita A, Obinata A, Hara N, Mitsumori R, Kaneda D, Hashizume Y, Sano T, Takao M, Gabdulkhaev R, Tada M, Kakita A, Arakawa A, Morishima M, Murayama S, Saito Y, Hatsuta H, Matsubara T, Akagi A, Riku Y, Miyahara H, Sone J, Yoshida M, Yamaguchi H, Tsukie T, Hasegawa M, Kasuga K, Kikuchi M, Akatsu H, Kuwano R, Iwatsubo T, Japanese Alzheimer's Disease Neuroimaging Initiative, Niida S, Ozaki K, Ikeuchi T. Association of rare APOE missense variants with Alzheimer's disease in the Japanese population. J Alzheimers Dis 2025:13872877251340710. [PMID: 40397079 DOI: 10.1177/13872877251340710] [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: 05/22/2025]
Abstract
BackgroundLittle is known about the rare missense variants (RMVs) of APOE in East Asians, including the Japanese, and their association with Alzheimer's disease (AD) and lipid metabolism.ObjectiveTo identify APOE RMVs in the Japanese population and investigate their association with AD and lipid metabolism, including low-density lipoprotein cholesterol levels.MethodsAPOE RMVs were explored in the Niigata (NIG; 2589 subjects) and Tohoku (ToMMo; 3307 subjects) cohorts. A case-control study included 6261 AD cases and 16,331 controls, all of whom were aged 65 or older. Sanger sequencing, whole-exome sequencing, or a combination of both was performed on the NIG subjects. We used the genotype data from the ToMMo cohort. APOE RMV frequencies in the Japanese population were compared with various ethnic populations. Associations between APOE RMV genotypes, AD, and lipoproteins were examined.ResultsFourteen RMVs were identified (minor allele frequency 0.02-0.73%), with 10 unique to East Asians. Five previously reported RMVs, such as the Christchurch RMV, were absent in Japanese individuals. Two RMVs (rs140808909 and rs190853081), which exhibit complete linkage disequilibrium, were found to have protective effects against AD: pBonferroni = 4.28E-02, OR (95% CI) = 0.70 (0.54-0.92). No significant differences in cholesterol levels were observed between RMV carriers and non-carriers.ConclusionsThe two APOE RMVs identified in Japanese individuals may have exhibited potential protective effects against AD. Further large-scale studies are needed to confirm these findings and to explore their roles in AD and lipid metabolism.
Collapse
Affiliation(s)
- Akinori Miyashita
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Ai Obinata
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Risa Mitsumori
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Daita Kaneda
- Department of Neuropathology, Fukushimura Hospital, Toyohashi, Japan
| | - Yoshio Hashizume
- Department of Neuropathology, Fukushimura Hospital, Toyohashi, Japan
| | - Terunori Sano
- Department of Clinical Laboratory and Internal Medicine, National Center of Neurology and Psychiatry (NCNP), National Center Hospital, Kodaira, Japan
| | - Masaki Takao
- Department of Clinical Laboratory and Internal Medicine, National Center of Neurology and Psychiatry (NCNP), National Center Hospital, Kodaira, Japan
| | - Ramil Gabdulkhaev
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Mari Tada
- Department of Pathology Neuroscience, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akira Arakawa
- Department of Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-ku, Japan
| | - Maho Morishima
- Department of Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-ku, Japan
| | - Shigeo Murayama
- Department of Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-ku, Japan
- Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Yuko Saito
- Department of Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-ku, Japan
| | - Hiroyuki Hatsuta
- Department of Neurology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
- Hatsuta Neurology Clinic, Osaka, Japan
| | - Tomoyasu Matsubara
- Department of Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi-ku, Japan
| | - Akio Akagi
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Yuichi Riku
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Hiroaki Miyahara
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Jun Sone
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Japan
| | | | - Tamao Tsukie
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Mai Hasegawa
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masataka Kikuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroyasu Akatsu
- Center for Frailty and Locomotive Syndrome, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Ryozo Kuwano
- Social Welfare Corporation Asahigawaso, Asahigawaso Research Institute, Okayama, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
- Director's Office, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Japan
- Japanese Alzheimer's Disease Neuroimaging Initiative, Japan
| | | | - Shumpei Niida
- Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
7
|
Hushmandi K, Lam HY, Wong WM, Tan W, Daryabari SH, Reiter RJ, Farahani N, Kumar AP. Gene therapy for age-related macular degeneration: a promising frontier in vision preservation. Cell Commun Signal 2025; 23:233. [PMID: 40394614 PMCID: PMC12090701 DOI: 10.1186/s12964-025-02246-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 05/11/2025] [Indexed: 05/22/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of central vision loss, progressively impairing the retina and affecting millions worldwide. By 2040, global cases of AMD are projected to reach 300 million, posing a significant public health challenge. While early AMD may only cause mild visual impairment, advanced stages, particularly neovascular (wet) and non-neovascular (dry) AMD, can lead to severe vision loss or legal blindness, substantially affecting daily life. The introduction of anti-angiogenic therapies has revolutionized wet AMD treatment, offering a high probability of preserving or improving vision. However, these therapies do not halt AMD progression, and no definitive treatments exist for dry AMD. The limitations of current therapies, such as frequent injections and treatment resistance, underscore the urgent need for novel strategies. Gene therapy, which has shown success in treating other hereditary retinal diseases, offers a promising long-term solution for AMD by targeting retinal cells to produce therapeutic proteins. This review explores the potential of gene therapy for AMD, examining recent clinical trials and future treatment directions.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- Yong Loo Lin School of Medicine, NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, 117599, Singapore
| | - Wendy Meihua Wong
- Centre for Innovation & Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Ophthalmology, National University Hospital, National University Health System, Singapore, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- Yong Loo Lin School of Medicine, NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, 117599, Singapore
- School of Chemical & Life Sciences, Singapore Polytechnic, 500 Dover Road, Singapore, 139651, Singapore
| | - Seyed-Hashem Daryabari
- Basir Eye Health Research Center, Tehran, Iran
- Trauma Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- Yong Loo Lin School of Medicine, NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
8
|
Sugandhi VV, Gadhave DG, Ugale AR, Kulkarni N, Nangare SN, Patil HP, Rath S, Saxena R, Lavate A, Patel AT, Jadhav A, Paudel KR. Advances in Alzheimer's Therapy: Exploring Neuropathological Mechanisms to Revolutionize the Future Therapeutic Landscape. Ageing Res Rev 2025; 109:102775. [PMID: 40403980 DOI: 10.1016/j.arr.2025.102775] [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: 12/12/2024] [Revised: 05/07/2025] [Accepted: 05/16/2025] [Indexed: 05/24/2025]
Abstract
Alzheimer's disease (AD) is still an excessively complicated neurological disorder that impacts millions of individuals globally. The ideal defensive feature of the central nervous system (CNS) is the intimate junction of endothelial cells, which functions as a biological barrier to safely control molecular transport throughout the brain. The blood-brain barrier (BBB) comprises tightly locked astrocyte cell junctions on CNS blood capillaries. This biological barrier shields the brain from hazardous toxins by preventing the entry of polar medications, cells, and ions. However, it is very challenging to provide any treatment to the brain for neurodegenerative illnesses like Alzheimer's. Different causative mechanisms, such as amyloid-β (Aβ) plaques, tubulin-associated unit (Tau) tangles, and neuroinflammation, cause neuronal dysfunction, leading to dementia and memory loss in the subject. Several treatments are approved for AD therapy, whereas most only help treat related symptoms. Disappointingly, current remedies have not been able to control the progression of AD due to associated side effects. Specific pathogenic mechanisms are involved in the initiation and development of this disease. Therefore, the expected survival of a patient with AD is limited and is approximately ten years. Hence, the pathogenic mechanism behind AD progression must be understood to better comprehend and improve the overall survival rate. This review highlighted the recent insights into AD pathogenesis, molecular mechanisms, advancements in theragnostic techniques, the existing updates of clinical trials, and emerging innovations for AD medicinal development. That has helped researchers develop other strategies to address the shortcomings of traditional medications.
Collapse
Affiliation(s)
- Vrashabh V Sugandhi
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Dnyandev G Gadhave
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA; Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune, Maharashtra 413130, India.
| | - Akanksha R Ugale
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Nilesh Kulkarni
- Department of Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Sopan N Nangare
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Harshal P Patil
- Department of Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Seepra Rath
- Center for Dermal Research, Rutgers, the State University of New Jersey, 145 Bevier Road, Piscataway 08854, USA
| | - Rahul Saxena
- Graduate Programs in Molecular Biosciences, Rutgers, the State University of New Jersey, Piscataway 08854, USA
| | - Amol Lavate
- Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune, Maharashtra 413130, India
| | - Apeksha T Patel
- Department of Quality Assurance, Navinta III INC, Boca Raton, 33487, Florida, USA
| | - Ashish Jadhav
- Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune, Maharashtra 413130, India
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Science, Centenary Institute and University of Technology Sydney, Sydney 2007, Australia.
| |
Collapse
|
9
|
Aathira NS, Kaur A, Kumar A, Dar GM, Nimisha, Sharma AK, Bera P, Mahajan B, Chatterjee A, Saluja SS. The genetic risk factors, molecular pathways, microRNAs, and the gut microbiome in Alzheimer's disease. Neuroscience 2025; 577:217-227. [PMID: 40374065 DOI: 10.1016/j.neuroscience.2025.05.021] [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: 11/23/2024] [Revised: 04/25/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia worldwide. It is a multifaceted condition resulting from interplay of genetic mutations (e.g., APP, PSEN1, PSEN2) that account for less than 5% of cases, several genetic risk variants such as APOE4, TREM2, CD33, CLU, SORL1, and CR1 contribute to disease susceptibility and epigenetic factors, which may mediate the influence of environmental and lifestyle factors over time. Other critical contributors such as aging, protein misfolding and aggregation (amyloid-β and tau), molecular and transcriptomic dysregulation affecting neuronal function, and modifiable lifestyle factors like diet, physical activity, and environmental exposures presents challenges in accurate diagnosis and management. Research has predominantly focused on the diverse molecular pathways in the pathogenesis of AD, with particular attention given to the amyloidogenic pathways, tau pathology, calcium signalling, endolysosomal pathways, and others, whether they are directly or indirectly involved. Apart from these known molecular pathways, miRNAs are gaining attention as important regulators, which have been implicated in moderating the expression of mRNA targets involved in various processes associated with the clearance of pathogenic β-amyloid proteins. A mounting body of research suggests the possible role of gut microbiota in AD which regulates inflammation, neurotransmitters, and the blood-brain barrier. Gut dysbiosis can trigger neuroinflammation and amyloid-beta aggregation, making microbiome composition a potential early AD biomarker. This review aims to explore briefly the diverse risk encompassing genetic polymorphisms, altered molecular pathways implicated in AD pathogenesis, miRNA regulatory mechanisms, and the potential impact of gut microbiota on AD risk.
Collapse
Affiliation(s)
- N S Aathira
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Amanpreet Kaur
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Ghulam Mehdi Dar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Abhay Kumar Sharma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Pinki Bera
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Bhawna Mahajan
- Department of Biochemistry, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Atri Chatterjee
- Department of Neurology, VMMC and Safdarjung Hospital, New Delhi, India.
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of GI Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India.
| |
Collapse
|
10
|
Awan MUN, Mahmood F, Peng XB, Zheng F, Xu J. Underestimated virus impaired cognition-more evidence and more work to do. Front Immunol 2025; 16:1550179. [PMID: 40421014 PMCID: PMC12104254 DOI: 10.3389/fimmu.2025.1550179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Neurodegenerative disorders (NDs) are chronic neurological diseases that can be of idiopathic, genetic, or potentially infectious origin. Although the exact cause of neurodegeneration is unknown, it might be result of a confluence of age, genetic susceptibility factors, and environmental stresses. The blood-brain barrier shields the brain from the majority of viral infections, however neurotropic viruses are able to breach this barrier and infect central nervous system. Growing research points to a possible connection between viruses and neurodegenerative diseases, indicating that virus-induced neuroinflammation and disruption of neuronal protein quality control may play a role in the initial stages of disease progression. The diagnosis and treatment of NDs are urgent and challenging. Even though there is limited clinical evidence to support the use of antiviral medications and their dose regimens within the central nervous system (CNS), with the exception of acyclovir, they are currently utilized to treat various viral CNS infections. Understanding the neuropathogenesis of viral CNS infection may help with targeted diagnosis and treatment plans by focusing on the molecular mechanisms of the CNS. It may also be helpful in the search for new antiviral drugs, which are crucial for better managing these neurotropic viral infections. This review focuses on new findings linking viral infection to NDs and explores how viral modifications of cellular functions can impact the development of neurodegeneration and will also explore the therapeutic potential of antiviral drugs in NDs.
Collapse
Affiliation(s)
- Maher Un Nisa Awan
- Department of Neurology, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Faisal Mahmood
- Central Laboratory, Liver Disease Research Center and Department of Infectious Disease, The Affiliated Hospital of Yunnan University, Kunming, China
| | - Xiao-bin Peng
- Department of Neurology, The Affiliated Hospital of Yunnan University, Kunming, China
- School of Medicine, Yunnan University, Kunming, Yunnan, China
| | - Fenshuang Zheng
- Department of Emergency, The Affiliated hospital of Yunnan University, Kunming, China
| | - Jun Xu
- Department of Neurology, The Affiliated Hospital of Yunnan University, Kunming, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Lwere K, Muwonge H, Sendagire H, Sajatovic M, Gumukiriza-Onoria JL, Buwembo D, Buwembo W, Nassanga R, Nakimbugwe R, Nazziwa A, Munabi IG, Nakasujja N, Kaddumukasa M. Apolipoprotein E (APOE) and Alzheimer's disease risk in a Ugandan population: A pilot case-control study. Medicine (Baltimore) 2025; 104:e42407. [PMID: 40355218 PMCID: PMC12074064 DOI: 10.1097/md.0000000000042407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by cognitive decline and progressive functional impairment. The Apolipoprotein E (APOE) gene, particularly its ε2, ε3, and ε4 alleles, plays a crucial role in lipid metabolism, and has been implicated in AD pathogenesis. Although the APOE ε4 status is associated with an increased risk of AD, its impact varies across populations. This study investigated the prevalence of and association between APOE alleles and AD risk in a Ugandan cohort. This case-control study was conducted in Uganda, and included 87 participants (45 patients with AD and 42 healthy controls). Cognitive assessment was performed using the Montreal Cognitive Assessment (MoCA) and clinical diagnoses were based on the ICD-11 and DSM-5 criteria. Venous blood was collected for APOE genotyping by polymerase chain reaction. Statistical analyses, including logistic regression and generalized additive models (GAMs), were used to assess the association between APOE alleles and AD risk after adjusting for age, education, and sex. This study included 45 patients with AD and 42 healthy controls. The AD group was significantly older than controls (79.6 vs 73.0 years; P = .0006). The ε4 allele was common in both the AD (42.2%) and control groups (44.0%), which was higher than the 1000 Genomes African ancestry data. No significant association was found between the APOE genotype or allele dosage and AD risk after adjusting for age, sex, and education. However, the probability of AD increases with age, particularly among ε4 carriers with lower educational levels. While APOE ε4 status was associated with a higher predicted probability of AD in older adults, no statistically significant relationship was observed in the Ugandan cohort. These findings support the need for larger population-specific studies to explore APOE's role of APOE in AD risk across sub-Saharan Africa.
Collapse
Affiliation(s)
- Kamada Lwere
- Department of Microbiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
- Department of Microbiology, Faculty of Health Sciences, Soroti University, Soroti, Uganda
- Habib Medical School, Faculty of Health Sciences, Islamic University in Uganda, Kampala, Uganda
| | - Haruna Muwonge
- Department of Physiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Hakim Sendagire
- Department of Microbiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Martha Sajatovic
- Neurological and Behavioral Outcomes Center, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH
| | | | - Denis Buwembo
- School of Public Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | - William Buwembo
- Department of Anatomy, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Rita Nassanga
- Department of Radiology, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Rheem Nakimbugwe
- Habib Medical School, Faculty of Health Sciences, Islamic University in Uganda, Kampala, Uganda
| | - Aisha Nazziwa
- Habib Medical School, Faculty of Health Sciences, Islamic University in Uganda, Kampala, Uganda
| | - Ian Guyton Munabi
- Department of Anatomy, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Noeline Nakasujja
- Department of Psychiatry, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Mark Kaddumukasa
- Department of Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
12
|
Chen JH, Chan L, Hong CT, Hu CJ, Hsieh YC. Role of apolipoprotein E (ApoE) ε4 in cognitive impairment after a stroke: a prospective cohort study. Aging (Albany NY) 2025; 17:206248. [PMID: 40349252 DOI: 10.18632/aging.206248] [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: 01/20/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025]
Abstract
Although apolipoprotein E (ApoE) ε4 is a well-established risk factor for Alzheimer disease, its role in the development of post-stroke cognitive impairment (PSCI) remains uncertain. In this prospective cohort study, we recruited patients aged ≥20 years who had ischemic stroke within the past 7 days and measured their ApoE genotype. Baseline characteristics, including age, sex, education level, medical history, stroke severity, stroke etiology, and neuroimaging findings were recorded. Cognitive function was evaluated using the Montreal Cognitive Assessment (MoCA) and Clinical Dementia Rating (CDR) at 3 and 12 months post-stroke, with PSCI defined as a MoCA score < 26. After adjusting for confounding factors, the ApoE ε4 allele was not associated with the risk of PSCI at 3 or 12 months post-stroke. Other factors, including age, body mass index, education level, and initial stroke severity, were found to be associated with the risk of PSCI at 3 months. In patients who developed PSCI at 12 months, only education level and the MoCA score at 3 months were significantly associated with the risk of PSCI. Our findings suggest that, aside from traditional risk factors, the ApoE ε4 allele does not contribute to the risk of PSCI at 3 or 12 months post-stroke. Further studies with a larger sample size and longer follow-up are warranted.
Collapse
Affiliation(s)
- Jia-Hung Chen
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei, Taiwan
| | - Lung Chan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei, Taiwan
| | - Chien-Tai Hong
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei, Taiwan
| | - Chaur-Jong Hu
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, New Taipei, Taiwan
| | - Yi-Chen Hsieh
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
13
|
Li Z, Martens YA, Ren Y, Jin Y, Sekiya H, Doss SV, Kouri N, Castanedes-Casey M, Christensen TA, Miller Nevalainen LB, Takegami N, Chen K, Liu CC, Soto-Beasley A, Boon BDC, Labuzan SA, Ikezu TC, Chen Y, Bartkowiak AD, Xhafkollari G, Wetmore AM, Bennett DA, Reichard RR, Petersen RC, Kanekiyo T, Ross OA, Murray ME, Dickson DW, Bu G, Zhao N. APOE genotype determines cell-type-specific pathological landscape of Alzheimer's disease. Neuron 2025; 113:1380-1397.e7. [PMID: 40112813 DOI: 10.1016/j.neuron.2025.02.017] [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: 01/17/2024] [Revised: 11/21/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
The apolipoprotein E (APOE) gene is the strongest genetic risk modifier for Alzheimer's disease (AD), with the APOE4 allele increasing risk and APOE2 decreasing it compared with the common APOE3 allele. Using single-nucleus RNA sequencing of the temporal cortex from APOE2 carriers, APOE3 homozygotes, and APOE4 carriers, we found that AD-associated transcriptomic changes were highly APOE genotype dependent. Comparing AD with controls, APOE2 carriers showed upregulated synaptic and myelination-related pathways, preserving synapses and myelination at the protein level. Conversely, these pathways were downregulated in APOE3 homozygotes, resulting in reduced synaptic and myelination proteins. In APOE4 carriers, excitatory neurons displayed reduced synaptic pathways similar to APOE3, but oligodendrocytes showed upregulated myelination pathways like APOE2. However, their synaptic and myelination protein levels remained unchanged or increased. APOE4 carriers also showed increased pro-inflammatory signatures in microglia but reduced responses to amyloid-β pathology. These findings reveal APOE genotype-specific molecular alterations in AD across cell types.
Collapse
Affiliation(s)
- Zonghua Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yunjung Jin
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Hiroaki Sekiya
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Sydney V Doss
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Naomi Kouri
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | | | - Nanaka Takegami
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Kai Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Baayla D C Boon
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Sydney A Labuzan
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tadafumi C Ikezu
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | - Allison M Wetmore
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ross R Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
14
|
Lee S, Kelly RS, Chen Y, Waqas M, Mendez KM, Hecker J, Hahn G, Lutz SM, Celedón JC, Clish CB, Litonjua AA, Chen Q, McGeachie M, Choi Y, Weiss ST, Tanzi RE, Lange C, Prokopenko D, Lasky-Su JA. Associations of APOE variants with sphingomyelin and cholesterol metabolites across the life-course in diverse populations. Metabolomics 2025; 21:64. [PMID: 40335834 DOI: 10.1007/s11306-025-02256-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 04/02/2025] [Indexed: 05/09/2025]
Abstract
INTRODUCTION Two alleles (ε2 and ε4) in the APOE gene are known to be strongly associated with lipid metabolism disorders, such as dyslipidemia, which are important risk factors for the development of cardiovascular diseases. While prior research has largely centered on adult populations, establishing APOE-lipid associations in infants, children, and adolescents-especially those from historically understudied groups-could inform earlier interventions and treatments. OBJECTIVES This study aimed to evaluate the dependence of the metabolome on the APOE variants using five diverse cohorts that span infancy through adulthood, comprising a total of over 190,000 individuals. METHODS We extracted the APOE variants (rs7412 and rs429358) from all cohorts-testing both the ε2 allele (rs7412-T and rs429358-T) and the ε4 allele (rs7412-C and rs429358-C)-and evaluated their associations with the global plasma metabolome which was measured by mass spectrometry-based (Metabolon or Broad Institute) or NMR-based (Nightingale) assays depending on the cohort, using a Bonferroni-corrected significance threshold. RESULTS Among 589 metabolites tested in our discovery population, only six including sphingomyelins and cholesterol were significantly associated with the rs7412/ε2 allele. Sphingomyelin (d18:1/22:0) and cholesterol were negatively associated with ε2 allele (β-value = -0.54 [-0.76, -0.32] p-value = 1.39 × 10-6 and - 0.55 [-0.77, -0.33]; p-value = 1.49 × 10-6, respectively). These relationships were replicated in the four additional cohorts without heterogeneity. CONCLUSION Our findings support the need for early intervention in lipid levels regardless of age, sex, and ethnicity and further investigations of the APOE variants on risk of various diseases in later life.
Collapse
Grants
- R01HL169300 the National Heart, Lung, and Blood Institute
- R01HL169300 the National Heart, Lung, and Blood Institute
- R01HL169300 the National Heart, Lung, and Blood Institute
- R01MH129337 the National Heart, Lung, and Blood Institute
- P01 HL132825 NHLBI NIH HHS
- R01HL169300 the National Heart, Lung, and Blood Institute
- R01HL169300 the National Heart, Lung, and Blood Institute
Collapse
Affiliation(s)
- Sanghun Lee
- Department of Medical Consilience, Division of Medicine, Graduate School, Dankook University, Yongin-si, South Korea
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rachel S Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Yulu Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mohammad Waqas
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Kevin M Mendez
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Julian Hecker
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Georg Hahn
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sharon M Lutz
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Healthcare Institute, Boston, MA, USA
| | - Juan C Celedón
- Division of Pediatric Pulmonary, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Clary B Clish
- Metabolomics Platform, Broad Institute, Cambridge, MA, USA
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children's Hospital at Strong, University of Rochester Medical Center, Rochester, NY, USA
| | - Qingwen Chen
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Younjung Choi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Christoph Lange
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Dmitry Prokopenko
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
15
|
Liu S, Feng A, Li Z. Neuron-Derived Extracellular Vesicles: Emerging Regulators in Central Nervous System Disease Progression. Mol Neurobiol 2025:10.1007/s12035-025-05010-4. [PMID: 40325332 DOI: 10.1007/s12035-025-05010-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
The diagnosis and exploration of central nervous system (CNS) diseases remain challenging due to the blood-brain barrier (BBB), complex signaling pathways, and heterogeneous clinical manifestations. Neurons, as the core functional units of the CNS, play a pivotal role in CNS disease progression. Extracellular vesicles (EVs), capable of crossing the BBB, facilitate intercellular and cell-extracellular matrix (ECM) communication, making neuron-derived extracellular vesicles (NDEVs) a focal point of research. Recent studies reveal that NDEVs, carrying various bioactive substances, can exert either pathogenic or protective effects in numerous CNS diseases. Additionally, NDEVs show significant potential as biomarkers for CNS diseases. This review summarizes the emerging roles of NDEVs in CNS diseases, including Alzheimer's disease, depression, traumatic brain injury, schizophrenia, ischemic stroke, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. It aims to provide a novel perspective on developing therapeutic and diagnostic strategies for CNS diseases through the study of NDEVs.
Collapse
Affiliation(s)
- Sitong Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Aitong Feng
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Zhigang Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, 518107, China.
| |
Collapse
|
16
|
Zhao Q, Xu J, Shi Z, Zhang Y, Du X, Zhai Y, Xu J, Liu F, Zhang Q. Genome-wide Pleiotropy Analysis Reveals Shared Genetic Associations between Type 2 Diabetes Mellitus and Subcortical Brain Volumes. RESEARCH (WASHINGTON, D.C.) 2025; 8:0688. [PMID: 40330659 PMCID: PMC12053431 DOI: 10.34133/research.0688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025]
Abstract
Type 2 diabetes mellitus (T2DM), a prevalent metabolic disorder marked by insulin resistance and hyperglycemia, has been linked to volumetric changes in subcortical regions, yet the genetic basis of this relationship remains unclear. We analyzed genome-wide association study summary data for T2DM and 14 subcortical volumetric traits, using MiXeR to quantify shared genetic architecture and applying conditional/conjunctional false discovery rate analyses to detect novel and shared genomic loci. Enrichment and gene expression analyses were subsequently performed to explore the biological functions and mechanisms of genes associated with these loci. We observed a substantial proportion of trait-influencing variants shared between T2DM and subcortical structures, with Dice coefficients ranging from 22.4% to 49.6%. Additionally, 70 distinct loci were identified as being jointly associated with T2DM and subcortical volumes, 5 and 22 of which were novel for T2DM and subcortical volumes, respectively. The 769 protein-coding genes mapped to these shared loci are enriched in metabolic and neurodevelopmental pathways and exhibit specific developmental trajectories, with 117 genes showing expression levels linked to both T2DM and subcortical structures. This study uncovered polygenic overlap between T2DM and subcortical structures, deepening our comprehension of the genetic factors linking metabolic disorders and brain health.
Collapse
Affiliation(s)
| | | | | | - Yang Zhang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology,
Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Du
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology,
Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ying Zhai
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology,
Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jinglei Xu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology,
Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology,
Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Quan Zhang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology,
Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
17
|
Timofeeva AM, Aulova KS, Nevinsky GA. Modeling Alzheimer's Disease: A Review of Gene-Modified and Induced Animal Models, Complex Cell Culture Models, and Computational Modeling. Brain Sci 2025; 15:486. [PMID: 40426657 PMCID: PMC12109626 DOI: 10.3390/brainsci15050486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/30/2025] [Accepted: 05/03/2025] [Indexed: 05/29/2025] Open
Abstract
Alzheimer's disease, a complex neurodegenerative disease, is characterized by the pathological aggregation of insoluble amyloid β and hyperphosphorylated tau. Multiple models of this disease have been employed to investigate the etiology, pathogenesis, and multifactorial aspects of Alzheimer's disease and facilitate therapeutic development. Mammals, especially mice, are the most common models for studying the pathogenesis of this disease in vivo. To date, the scientific literature has documented more than 280 mouse models exhibiting diverse aspects of Alzheimer's disease pathogenesis. Other mammalian species, including rats, pigs, and primates, have also been utilized as models. Selected aspects of Alzheimer's disease have also been modeled in simpler model organisms, such as Drosophila melanogaster, Caenorhabditis elegans, and Danio rerio. It is possible to model Alzheimer's disease not only by creating genetically modified animal lines but also by inducing symptoms of this neurodegenerative disease. This review discusses the main methods of creating induced models, with a particular focus on modeling Alzheimer's disease on cell cultures. Induced pluripotent stem cell (iPSC) technology has facilitated novel investigations into the mechanistic underpinnings of diverse diseases, including Alzheimer's. Progress in culturing brain tissue allows for more personalized studies on how drugs affect the brain. Recent years have witnessed substantial advancements in intricate cellular system development, including spheroids, three-dimensional scaffolds, and microfluidic cultures. Microfluidic technologies have emerged as cutting-edge tools for studying intercellular interactions, the tissue microenvironment, and the role of the blood-brain barrier (BBB). Modern biology is experiencing a significant paradigm shift towards utilizing big data and omics technologies. Computational modeling represents a powerful methodology for researching a wide array of human diseases, including Alzheimer's. Bioinformatic methodologies facilitate the analysis of extensive datasets generated via high-throughput experimentation. It is imperative to underscore the significance of integrating diverse modeling techniques in elucidating pathogenic mechanisms in their entirety.
Collapse
Affiliation(s)
- Anna M. Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk 630090, Russia
| | | | | |
Collapse
|
18
|
Lozupone M, Dibello V, Resta E, Sardone R, Castellana F, Zupo R, Lampignano L, Bortone I, Mollica A, Berardino G, Altamura M, Bellomo A, Daniele A, Solfrizzi V, Panza F. Uncertainties in anti-amyloid monoclonal antibody therapy for Alzheimer's disease: the challenges ahead. Expert Rev Neurother 2025:1-11. [PMID: 40314575 DOI: 10.1080/14737175.2025.2500752] [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: 02/09/2025] [Revised: 04/04/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD), the leading cause of dementia, poses a significant burden on patients, caregivers, and healthcare systems worldwide. After two decades of extensive efforts, we are still without significantly effective disease-modifying drugs for AD. Although brain amyloid-β (Aβ) accumulation may predict cognitive decline, several drug candidates, including anti-Aβ monoclonal antibodies, have been developed and tested to reduce Aβ plaque burden effective, but without significant clinical success. AREAS COVERED The following review presents and discusses anti-Aβ monoclonal antibody therapeutics used to treat AD. The article considers both current approaches and alternatives. This article is multiple database searches (MEDLINE, EMBASE, Scopus, Ovid and Google Scholar) on all the available literature up to 1 February 2025. EXPERT OPINION Randomized clinical trials (RCTs) of anti-Aβ drugs in AD have not fully validated the Aβ cascade hypothesis. Nevertheless, eight anti-Aβ monoclonal antibodies have, thus far, made it to Phase III RCTs. Moving forward, the use of the Apolipoprotein E genotype and tau protein as alternative biomarkers can assist clinicians in providing patients with even more individualized and efficacious anti-Aβ monoclonal antibodies dosing regimens and reduce the risk of serious amyloid-related imaging abnormalities.
Collapse
Affiliation(s)
- Madia Lozupone
- Department of Translational Biomedicine and Neuroscience "DiBrain", University of Bari Aldo Moro, Bari, Italy
| | - Vittorio Dibello
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Emanuela Resta
- Translational Medicine and Health System Management, Department of Economy, University of Foggia, Foggia, Italy
| | - Rodolfo Sardone
- Unit of Statistics and Epidemiology, Local Healthcare Authority of Taranto, Taranto, Italy
| | - Fabio Castellana
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| | - Roberta Zupo
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| | - Luisa Lampignano
- Unit of Statistics and Epidemiology, Local Healthcare Authority of Taranto, Taranto, Italy
| | - Ilaria Bortone
- Local Healthcare Authority of Bari, ASL Bari, Bari, Italy
| | - Anita Mollica
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giuseppe Berardino
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Mario Altamura
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical & Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonio Daniele
- Department of Neuroscience, Catholic University of Sacred Heart, Rome, Italy
- Neurology Unit, IRCCS Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - Vincenzo Solfrizzi
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Panza
- Department of Interdisciplinary Medicine (DIM), "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
19
|
Ossenkoppele R, Salvadó G, Janelidze S, Pichet Binette A, Bali D, Karlsson L, Palmqvist S, Mattsson-Carlgren N, Stomrud E, Therriault J, Rahmouni N, Rosa-Neto P, Coomans EM, van de Giessen E, van der Flier WM, Teunissen CE, Jonaitis EM, Johnson SC, Villeneuve S, Benzinger TLS, Schindler SE, Bateman RJ, Doecke JD, Doré V, Feizpour A, Masters CL, Rowe C, Wiste HJ, Petersen RC, Jack CR, Hansson O. Plasma p-tau217 and tau-PET predict future cognitive decline among cognitively unimpaired individuals: implications for clinical trials. NATURE AGING 2025; 5:883-896. [PMID: 40155777 DOI: 10.1038/s43587-025-00835-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 02/12/2025] [Indexed: 04/01/2025]
Abstract
Plasma p-tau217 and tau positron emission tomography (PET) are strong prognostic biomarkers in Alzheimer's disease (AD), but their relative performance in predicting future cognitive decline among cognitively unimpaired (CU) individuals is unclear. In a head-to-head comparison study including nine cohorts and 1,474 individuals, we show that plasma p-tau217 and medial temporal lobe tau-PET signal display similar associations with cognitive decline on a global cognitive composite test (R2PET = 0.34 versus R2plasma = 0.33, Pdifference = 0.653) and with progression to mild cognitive impairment (hazard ratio (HR)PET = 1.61 (1.48-1.76) versus HRplasma = 1.57 (1.43-1.72), Pdifference = 0.322). Combined plasma and PET models were superior to the single-biomarker models (R2 = 0.35, P < 0.01). Sequential selection using plasma phosphorylated tau at threonine 217 (p-tau217) and then tau-PET reduced the number of participants required for a clinical trial by 94%, compared to a 76% reduction when using plasma p-tau217 alone. Thus, plasma p-tau217 and tau-PET showed similar performance for predicting future cognitive decline in CU individuals, and their sequential use enhances screening efficiency for preclinical AD trials.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands.
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Divya Bali
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Linda Karlsson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Research Centre for Studies in Aging, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Emma M Coomans
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Elsmarieke van de Giessen
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Department of Laboratory Medicine, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sylvia Villeneuve
- Centre for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- The Tracy Family SILQ Center, Washington University School of Medicine, St. Louis, MO, USA
| | - James D Doecke
- Australian eHealth Research Centre, Commonwealth Scientific and Industrial Research Organization, Melbourne, Victoria, Australia
| | - Vincent Doré
- Australian eHealth Research Centre, Commonwealth Scientific and Industrial Research Organization, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
| | - Azadeh Feizpour
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia
| | - Christopher Rowe
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Victoria, Australia
| | - Heather J Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences in Malmö, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
20
|
Wen H, He Y, Tang Y, Zhu L, Tao Q, Jin B, Luo T, Peng Y, Wei Y, Lei J, Wang L, Wang F, Ling F, Gao Y, Han L. Altered immune response is associated with sex difference in vulnerability to Alzheimer's disease in human prefrontal cortex. Brain Pathol 2025; 35:e13318. [PMID: 39497354 PMCID: PMC11961208 DOI: 10.1111/bpa.13318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/17/2024] [Indexed: 04/03/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with a higher risk incidence in females than in males, and there are also differences in AD pathophysiology between sexes. The role of sex in the pathogenesis of AD may be crucial, yet the cellular and molecular basis remains unclear. Here, we performed a comprehensive analysis using four public transcriptome datasets of AD patients and age-matched control individuals in prefrontal cortex, including bulk transcriptome (295 females and 402 males) and single-nucleus RNA sequencing (snRNA-seq) data (224 females and 219 males). We found that the transcriptomic profile in female control was similar to those in AD. To characterize the key features associated with both the pathogenesis of AD and sex difference, we identified a co-expressed gene module that positively correlated with AD, sex, and aging, and was also enriched with immune-associated pathways. Using snRNA-seq datasets, we found that microglia (MG), a resident immune cell in the brain, demonstrated substantial differences in several aspects between sexes, such as an elevated proportion of activated MG, altered transcriptomic profile and cell-cell interaction between MG and other brain cell types in female control. Additionally, genes upregulated in female MG, such as TLR2, MERTK, SPP1, SLA, ACSL1, and FKBP5, had high confidence to be identified as biomarkers to distinguish AD status, and these genes also interacted with some approved drugs for treatment of AD. These findings underscore the altered immune response in female is associated with sex difference in susceptibility to AD, and the necessity of considering sex factors when developing AD biomarkers and therapeutic strategies, providing a scientific basis for further in-depth studies on sex differences in AD.
Collapse
Affiliation(s)
- Huiying Wen
- BGI ResearchHangzhouChina
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
- BGI ResearchShenzhenChina
| | - Youzhe He
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Yuanchun Tang
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Langjian Zhu
- BGI ResearchHangzhouChina
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
- BGI ResearchShenzhenChina
| | - Quyuan Tao
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Bufan Jin
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Ting Luo
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Yujie Peng
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Yanrong Wei
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Junjie Lei
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Lifang Wang
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Fan Wang
- Department of Pathology of Sir Run Run Shaw Hospital, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
- Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
| | - Fei Ling
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
| | - Yue Gao
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- Department of Pathology of Sir Run Run Shaw Hospital, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
- Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
| | - Lei Han
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| |
Collapse
|
21
|
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
|
22
|
Cui Y, Ma X, Wei J, Chen C, Shakir N, Guirram H, Dai Z, Anderson T, Ferguson D, Qiu S. MET receptor tyrosine kinase promotes the generation of functional synapses in adult cortical circuits. Neural Regen Res 2025; 20:1431-1444. [PMID: 39075910 PMCID: PMC11624886 DOI: 10.4103/nrr.nrr-d-23-01471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/02/2024] [Accepted: 04/20/2024] [Indexed: 07/31/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202505000-00026/figure1/v/2024-07-28T173839Z/r/image-tiff Loss of synapse and functional connectivity in brain circuits is associated with aging and neurodegeneration, however, few molecular mechanisms are known to intrinsically promote synaptogenesis or enhance synapse function. We have previously shown that MET receptor tyrosine kinase in the developing cortical circuits promotes dendritic growth and dendritic spine morphogenesis. To investigate whether enhancing MET in adult cortex has synapse regenerating potential, we created a knockin mouse line, in which the human MET gene expression and signaling can be turned on in adult (10-12 months) cortical neurons through doxycycline-containing chow. We found that similar to the developing brain, turning on MET signaling in the adult cortex activates small GTPases and increases spine density in prefrontal projection neurons. These findings are further corroborated by increased synaptic activity and transient generation of immature silent synapses. Prolonged MET signaling resulted in an increased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid/N-methyl-D-aspartate (AMPA/NMDA) receptor current ratio, indicative of enhanced synaptic function and connectivity. Our data reveal that enhancing MET signaling could be an interventional approach to promote synaptogenesis and preserve functional connectivity in the adult brain. These findings may have implications for regenerative therapy in aging and neurodegeneration conditions.
Collapse
Affiliation(s)
- Yuehua Cui
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Xiaokuang Ma
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Jing Wei
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Chang Chen
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Neha Shakir
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Hitesch Guirram
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Zhiyu Dai
- Department of Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Trent Anderson
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Deveroux Ferguson
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| | - Shenfeng Qiu
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, USA
| |
Collapse
|
23
|
Yang X, Yao K, Zhang M, Zhang W, Zu H. New insight into the role of altered brain cholesterol metabolism in the pathogenesis of AD: A unifying cholesterol hypothesis and new therapeutic approach for AD. Brain Res Bull 2025; 224:111321. [PMID: 40164234 DOI: 10.1016/j.brainresbull.2025.111321] [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: 01/30/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
The dysregulation of cholesterol metabolism homeostasis has been universally suggested in the aeotiology of Alzheimer's disease (AD). Initially, studies indicate that alteration of serum cholesterol level might contribute to AD. However, because blood-brain barrier impedes entry of plasma cholesterol, brain cells are not directly influenced by plasma cholesterol. Furthermore, mounting evidences suggest a link between alteration of brain cholesterol metabolism and AD. Interestingly, Amyloid-β proteins (Aβ) can markedly inhibit cellular cholesterol biosynthesis and lower cellular cholesterol content in cultured cells. And Aβ overproduction/overload induces a significant decrease of brain cellular cholesterol content in familial AD (FAD) animals. Importantly, mutations or polymorphisms of genes related to brain cholesterol transportation, such as ApoE4, ATP binding cassette (ABC) transporters, low-density lipoprotein receptor (LDLR) family and Niemann-Pick C disease 1 or 2 (NPC1/2), obviously lead to decreased brain cholesterol transport, resulting in brain cellular cholesterol loss, which could be tightly associated with AD pathological impairments. Additionally, accumulating data show that there are reduction of brain cholesterol biosynthesis and/or disorder of brain cholesterol trafficking in a variety of sporadic AD (SAD) animals and patients. Collectively, compelling evidences indicate that FAD and SAD could share one common and overlapping neurochemical mechanism: brain neuronal/cellular cholesterol deficiency. Therefore, accumulated evidences strongly support a novel hypothesis that deficiency of brain cholesterol contributes to the onset and progression of AD. This review highlights the pivotal role of brain cholesterol deficiency in the pathogenesis of AD. The hypothesis offers valuable insights for the future development of AD treatment.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China; Department of Neurology, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200237, China
| | - Kai Yao
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Mengqi Zhang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Wenbin Zhang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China.
| |
Collapse
|
24
|
Tang JM, Lu Q, Lin HX, Li JQ, Zhu XC, Ma T. C-reactive protein-mediated dementia. Psychogeriatrics 2025; 25:e70032. [PMID: 40194896 DOI: 10.1111/psyg.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/18/2025] [Accepted: 03/27/2025] [Indexed: 04/09/2025]
Abstract
Loss of memory is the main feature of dementia, accompanied by personality changes. Alzheimer's disease (AD) is the most prevalent type of dementia globally and a major contributor to disability and mortality in older individuals. Most notably, the neurological damage caused by AD is irreversible, but the current market still lacks effective medications for the treatment of dementia. Numerous research studies have indicated that the inflammatory response is significantly involved in the development of cognitive impairment, and elevated C-reactive protein (CRP) levels in healthy people increases the likelihood of future AD. CRP is a nonspecific indicator of inflammation. In clinical practice, CRP has long been proven to be one of the risk factors and powerful predictors of neurodegenerative diseases. Given the accessibility and cost-effectiveness of CRP testing, it is reasonable to anticipate its utilisation for early screening and monitoring the progression of AD in the future. This review therefore focuses on the specific relationship between CRP and various types of dementia and explores how CRP contributes to cognitive impairment.
Collapse
Affiliation(s)
- Jia-Ming Tang
- Department of Neurology, the Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, China
| | - Qing Lu
- Department of Neurology, the Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, China
| | - Han-Xiao Lin
- Department of Neurology, the Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
| | - Jia-Qi Li
- Department of Neurology, the Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
| | - Xi-Chen Zhu
- Department of Neurology, the Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, China
- Department of Neurology, the Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
| | - Tao Ma
- Department of Neurology, the Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, China
- Department of Neurology, the Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, China
| |
Collapse
|
25
|
Sharma K, Rai P, Maurya SK, Tapadia MG. Anti-diabetic drug pioglitazone reduces Islet amyloid aggregation overload in the Drosophila neuronal cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6031-6041. [PMID: 39636405 DOI: 10.1007/s00210-024-03632-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
Amyloid-proteinopathy is observed in type 2 diabetes, where Islet amyloid polypeptide is secreted atypically and impedes cellular homeostasis. The thiazolidinediones family is reported to influence amyloid-beta aggregations. However, research on drug-based stimulation of insulin signaling to alleviate Islet amyloid aggregations is lacking. To understand the impact of pioglitazone on islet amyloid aggregation, we conducted an in vivo and in silico analysis. For in vivo analysis, we generated a transgenic Drosophila harboring the preproform of human Islet amyloid polypeptide (IAPP) that can be ectopically expressed in a spatio-temporal manner. We show that the unprocessed form of IAPP also has the propensity to form aggregates and cause degeneration. Pioglitazone feeding effectively reduces the burden of Islet amyloid aggregations in the larval brain. In silico analysis shows that there is a higher protein-ligand binding energy for IAPP with pioglitazone than amyloid-beta. These results suggests that pioglitazone might be repurposed as a drug to cure islet amyloidogenesis.
Collapse
Affiliation(s)
- Khushboo Sharma
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Pooja Rai
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India
| | - Madhu G Tapadia
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
26
|
Pu Y, Dai XL, Wang Y, Chen Y, Wu C, Zhou X, Chen M, Chen YH, Shuai X, Jin R, Nie Y. Hybrid Lipoplex Boosts Neuron-Microglia Crosstalk for Treatment of Alzheimer's Disease through Aβ-Targeted-Autophagy and ApoE2 Gene Supplementation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2418560. [PMID: 40296752 DOI: 10.1002/adma.202418560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/05/2025] [Indexed: 04/30/2025]
Abstract
Efficient clearance of amyloid-β (Aβ) is vital but challenging in Alzheimer's disease (AD) treatment due to its complicated regulation mechanisms during generation and metabolism. It necessitates a multidimensional synergistic strategy based on ingenious delivery system design. Herein, guanidine-rich lipids (metformin-inspired MLS and arginine-contained RLS) are devised to trigger selective chaperone-mediated autophagy for amyloid precursor protein degradation in neurons. They are further co-assembled with oleic acid-modified cerium dioxide (OA@CeO2) to form RMC assembly for pApoE2 delivery (RMC/pApoE2 lipoplex). The OA@CeO2 boosts macro-autophagy, alleviates oxidative stress and inflammatory microenvironment, and promotes the neurons-microglia crosstalk for Aβ elimination. Concurrently, both guanidine-rich lipids and OA@CeO2 benefit pApoE2 transfection in neurons, enabling the transport of Aβ into microglia, and facilitating enzymatic hydrolysis and cellular digestion of extracellular Aβ. The lipoplex-boosted neuron-microglia interactions ultimately eliminate both intra- and extra-cellular Aβ aggregates. Consequently, the RMC/pApoE2 lipoplex eliminates ≈86.9% of Aβ plaques in the hippocampus of APP/PS1 mice and restored the synaptic function and neuronal connectivity. Moreover, it recovers the spatial memory of APP/PS1 mice to nearly the level of WT control. The presented hybrid lipoplex showcases an advanced gene delivery system, and offers a promising strategy for Aβ clearance in AD treatment.
Collapse
Affiliation(s)
- Yiyao Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, P. R. China
| | - Xue-L Dai
- Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, 430072, P. R. China
| | - Yichun Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, P. R. China
| | - Yanbing Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, P. R. China
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, P. R. China
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu, Sichuan, 610065, P. R. China
| | - Xiangyu Zhou
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P. R. China
- Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, P. R. China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, SAR, 999078, P. R. China
| | - Yi-Hung Chen
- Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, 430072, P. R. China
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510630, P. R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan, 610064, P. R. China
| |
Collapse
|
27
|
Pardo-Pérez I, Sánchez-Valdeón L, García-Gallego A, Estébanez B, Casado-Verdejo I, Fernández-Fernández JA, Méndez-Martínez C, Bello-Corral L. Regional analysis of APOE polymorphism in Alzheimer's disease in Spain. Sci Rep 2025; 15:14877. [PMID: 40295636 PMCID: PMC12037765 DOI: 10.1038/s41598-025-98323-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with significant cognitive and functional impacts. Genetic factors, particularly the APOE gene and its allelic variants (ε2, ε3, ε4), play a critical role in AD susceptibility. This study analyzed the allelic frequency and distribution of APOE polymorphisms in three provinces of Castilla y León (León, Soria, Salamanca), Spain, to explore their potential relationship with AD risk. Genotypes were determined using polymerase chain reactions, and statistical analyses revealed significant regional variations. The ε3/ε3 genotype was the most prevalent overall, while the ε3/ε4 genotype predominated in specific areas like Ponferrada. The absence of homozygous ε4 individuals in Soria contrasts sharply with higher frequencies in Salamanca. These differences suggest historical and migratory influences on genetic variability. Identifying regional genetic patterns enhances our understanding of AD risk and supports the development of targeted preventive strategies. Early detection of high-risk alleles could improve patient outcomes, reduce healthcare burdens and inform public health policies.
Collapse
Affiliation(s)
| | | | - Ana García-Gallego
- Health Research Nursing Group (GREIS), University of León, Campus Vegazana, 24071, León, Spain
- Department of Economics and Statistics, University of León, 24071, León, Spain
| | - Brisamar Estébanez
- Institute of Biomedicine (IBIOMED), University of León, 24071, León, Spain
| | - Inés Casado-Verdejo
- Health Research Nursing Group (GREIS), University of León, Campus Vegazana, 24071, León, Spain
- Department of Nursing and Physiotherapy, University of León, 24401, Ponferrada, Spain
| | - Jesús Antonio Fernández-Fernández
- Department of Nursing and Physiotherapy, University of León, 24071, León, Spain
- Health Research Nursing Group (GREIS), University of León, Campus Vegazana, 24071, León, Spain
| | - Carlos Méndez-Martínez
- University Hospital of León, 24071, León, Spain
- Health Research Nursing Group (GREIS), University of León, Campus Vegazana, 24071, León, Spain
| | - Laura Bello-Corral
- Department of Nursing and Physiotherapy, University of León, 24401, Ponferrada, Spain
| |
Collapse
|
28
|
Hudák A, Letoha T. Endocytic Pathways Unveil the Role of Syndecans in the Seeding and Spreading of Pathological Protein Aggregates: Insights into Neurodegenerative Disorders. Int J Mol Sci 2025; 26:4037. [PMID: 40362276 PMCID: PMC12071627 DOI: 10.3390/ijms26094037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/07/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease and other neurodegenerative disorders are characterized by the accumulation of misfolded proteins, such as amyloid-beta, tau, and α-synuclein, which disrupt neuronal function and contribute to cognitive decline. Heparan sulfate proteoglycans, particularly syndecans, play a pivotal role in the seeding, aggregation, and spreading of toxic protein aggregates through endocytic pathways. Among these, syndecan-3 is particularly critical in regulating the internalization of misfolded proteins, facilitating their propagation in a prion-like manner. This review examines the mechanisms by which syndecans, especially SDC3, contribute to the seeding and spreading of pathological protein aggregates in neurodegenerative diseases. Understanding these endocytic pathways provides valuable insights into the potential of syndecans as biomarkers and therapeutic targets for early intervention in Alzheimer's disease and other related neurodegenerative disorders.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., 6726 Szeged, Hungary;
- Doctoral School of Theoretical Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Tamás Letoha
- Pharmacoidea Ltd., 6726 Szeged, Hungary;
- Doctoral School of Theoretical Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
29
|
Wendt S, Lin AJ, Ebert SN, Brennan DJ, Cai W, Bai Y, Kong DY, Sorrentino S, Groten CJ, Lee C, Frew J, Choi HB, Karamboulas K, Delhaye M, Mackenzie IR, Kaplan DR, Miller FD, MacVicar BA, Nygaard HB. A 3D human iPSC-derived multi-cell type neurosphere system to model cellular responses to chronic amyloidosis. J Neuroinflammation 2025; 22:119. [PMID: 40275379 PMCID: PMC12023538 DOI: 10.1186/s12974-025-03433-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by progressive amyloid beta (Aβ) deposition in the brain, with eventual widespread neurodegeneration. While the cell-specific molecular signature of end-stage AD is reasonably well characterized through autopsy material, less is known about the molecular pathways in the human brain involved in the earliest exposure to Aβ. Human model systems that not only replicate the pathological features of AD but also the transcriptional landscape in neurons, astrocytes and microglia are crucial for understanding disease mechanisms and for identifying novel therapeutic targets. METHODS In this study, we used a human 3D iPSC-derived neurosphere model to explore how resident neurons, microglia and astrocytes and their interplay are modified by chronic amyloidosis induced over 3-5 weeks by supplementing media with synthetic Aβ1 - 42 oligomers. Neurospheres under chronic Aβ exposure were grown with or without microglia to investigate the functional roles of microglia. Neuronal activity and oxidative stress were monitored using genetically encoded indicators, including GCaMP6f and roGFP1, respectively. Single nuclei RNA sequencing (snRNA-seq) was performed to profile Aβ and microglia driven transcriptional changes in neurons and astrocytes, providing a comprehensive analysis of cellular responses. RESULTS Microglia efficiently phagocytosed Aβ inside neurospheres and significantly reduced neurotoxicity, mitigating amyloidosis-induced oxidative stress and neurodegeneration following different exposure times to Aβ. The neuroprotective effects conferred by the presence of microglia was associated with unique gene expression profiles in astrocytes and neurons, including several known AD-associated genes such as APOE. These findings reveal how microglia can directly alter the molecular landscape of AD. CONCLUSIONS Our human 3D neurosphere culture system with chronic Aβ exposure reveals how microglia may be essential for the cellular and transcriptional responses in AD pathogenesis. Microglia are not only neuroprotective in neurospheres but also act as key drivers of Aβ-dependent APOE expression suggesting critical roles for microglia in regulating APOE in the AD brain. This novel, well characterized, functional in vitro platform offers unique opportunities to study the roles and responses of microglia to Aβ modelling key aspects of human AD. This tool will help identify new therapeutic targets, accelerating the transition from discovery to clinical applications.
Collapse
Affiliation(s)
- Stefan Wendt
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada.
| | - Ada J Lin
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Sarah N Ebert
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6 T 1Z4, Canada
| | - Declan J Brennan
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Wenji Cai
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Yanyang Bai
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Da Young Kong
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Stefano Sorrentino
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Christopher J Groten
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Christopher Lee
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Jonathan Frew
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Opalia Co, Montreal, QC, H2X 3Y7, Canada
| | - Hyun B Choi
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Konstantina Karamboulas
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, M5G 0 A4, Canada
| | - Mathias Delhaye
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Ian R Mackenzie
- Department of Pathology, Vancouver General Hospital, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - David R Kaplan
- Department of Medical Genetics, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, M5G 0 A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1 A8, Canada
| | - Freda D Miller
- Department of Medical Genetics, University of British Columbia, Vancouver, V6 T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6 T 1Z4, Canada
| | - Brian A MacVicar
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada
| | - Haakon B Nygaard
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6 T 1Z3, Canada.
| |
Collapse
|
30
|
Fan Y, Wang X, Ling Y, Wang Q, Zhou X, Li K, Zhou C. Identification and validation of biomarkers in Alzheimer's disease based on machine learning algorithms and single-cell sequencing analysis. Comput Biol Chem 2025; 118:108475. [PMID: 40315768 DOI: 10.1016/j.compbiolchem.2025.108475] [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: 12/05/2024] [Revised: 03/14/2025] [Accepted: 04/10/2025] [Indexed: 05/04/2025]
Abstract
OBJECTIVE Alzheimer's disease (AD) is a complicated neurodegenerative disease with unknown pathogenesis. Identifying possible diagnostic markers of AD is essential to elucidate its mechanisms and facilitate diagnosis. METHODS A total of 295 samples (153 AD and 142 normal) were analyzed from two datasets (GSE122063 and GSE132903) in the Gene Express Omnibus (GEO) database. Differentially expressed genes (DEGs) between groups were identified and dimensionality reduction was applied to identify feature genes (key genes) using three algorithms of machine learning including least absolute shrinkage and selection operator (LASSO), support vector machine-recursive feature elimination (SVM-RFE), and Random forest (RF). In addition, we obtained sample data from single-cell RNA datasets GSE157827, GSE167490, and GSE174367 to classify cells into different types and examined changes in gene expression and their correlation with AD progression. Immunofluorescence assay was used to verify the expression of key genes in animal experiments. RESULTS To identify diagnostic genes associated with AD, we analyzed two datasets and identified 379 DEGs which might be related to the onset of AD, and 115 of them were up-regulated and 264 down-regulated. Three algorithms of machine learning were adopted to reduce the dimensions of these DEGs and finally six core DEGs CD86, SCG3, VGF, PRKCG, SPP1, and TPI1 of AD were identified. Diagnostic analyses showed that SCG3 was substantially down-regulated in the AD group, and its AUC was higher in both the training and validation sets (0.845, 0.927, and 0.917, respectively). Transcriptome sequencing results further revealed that SCG3 expression was down-regulated in multiple cell types in the AD group and SCG3 expression in the hippocampus was found significantly reduced in the AD group. CONCLUSIONS This study systematically identified and validated the potential of SCG3 as an early diagnostic biomarker for AD through several technical strategies. The findings provided new biomarkers for early detection of AD and laid a foundation for future clinical applications.
Collapse
Affiliation(s)
- Yun Fan
- Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing 210046, China
| | - XiaoLong Wang
- Shuyang Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Shuyang 223600, China
| | - Yun Ling
- Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing 210046, China
| | - QiuYi Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - XiBin Zhou
- Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing 210046, China
| | - Kai Li
- Zhang Zhongjing Key Laboratory of Prescriptions and Immunomodulation, Zhang Zhongjing Traditional Chinese Medicine College, Nanyang Institute of Technologyinese Medicine, Nanyang 473000, China
| | - ChunXiang Zhou
- Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Nanjing 210046, China.
| |
Collapse
|
31
|
Li M, Wang Y, Kazis L, Xia W. Hypertension a Predictive Risk Factor on Progression to Alzheimer's Disease Using APOEε4 as a Benchmark. Brain Sci 2025; 15:434. [PMID: 40426605 PMCID: PMC12110230 DOI: 10.3390/brainsci15050434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/15/2025] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Comorbidities such as hypertension and hypercholesterolemia are risk factors associated with Mild Cognitive Impairment (MCI) and Alzheimer's disease (AD). The most significant genetic risk factor is the ε4 allele of the apolipoprotein E gene (APOE). The aim of this paper is to determine whether hypertension is the most significant but modifiable risk factor to delay AD onset. Method: A cohort of patients with MCI (N = 3052) is developed from the documented database (N = 43,999) within the National Alzheimer's Coordinating Center (NACC) during the time period from June 2005 to May 2021. Cox proportional hazard models with propensity score weights on demographic information and comorbidities at baseline are applied to examine association of hypertension and hypercholesterolemia with AD onset among MCI patients. Associations are compared to APOE genotypes and AD onset. In addition, the association of hypertension with decline rates in Mini-Mental State Examination (MMSE) scores are reported. Results: After controlling for age, sex, race, APOEε4, and reported comorbidities, the results show that MCI patients who subsequently develop hypertension within 18 months after their first diagnosis of MCI have a significantly higher risk of AD onset (HR = 2.77, 95%CI (1.66, 4.65), p value < 0.0001), compared to MCI patients with no hypertension or a late occurrence of hypertension after 18 months. This significant association is validated through a Random Forest method, a machine learning approach with bootstrap simulations. In addition, patients with early hypertension have significantly higher MMSE score declining rates compared to those without hypertension (coefficient = 0.988, p = 0.0054.). Conclusions: Hypertension is the most significant risk factor comparable to the genetic risk factor APOEε4 allele. Our finding is unique, as we did not observe a similar outcome in those with early hypercholesterolemia. Thus, among all comorbidities, hypertension is the most significant risk factor similar to the genetic risk factor APOEε4 allele.
Collapse
Affiliation(s)
- Mingfei Li
- Department of Mathematical Sciences, Bentley University, Waltham, MA 02452, USA
- Center for Healthcare Organization and Implementation Research, Bedford VA Healthcare System, Bedford, MA 01730, USA
| | - Ying Wang
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA 01730, USA;
- School of Computing and Data Science, Wentworth Institute of Technology, Boston, MA 02115, USA
| | - Lewis Kazis
- Department of Health Law, Policy and Management, Boston University School of Public Health, Boston, MA 02118, USA;
- Rehabilitation Outcomes Center (ROC), Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Weiming Xia
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA 01730, USA;
- Department of Biological Sciences, University of Massachusetts, Lowell, MA 01854, USA
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
32
|
Randhawa S, Saini TC, Bathla M, Bhardwaj R, Dhiman R, Acharya A. Nanomaterials in targeting amyloid-β oligomers: current advances and future directions for Alzheimer's disease diagnosis and therapy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:561-580. [PMID: 40297247 PMCID: PMC12035877 DOI: 10.3762/bjnano.16.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
The amyloid cascade hypothesis posits that amyloid-β oligomers (AβOs) are the most neurotoxic species in Alzheimer's disease (AD). These oligomers, characterized by their high β-sheet content, have been shown to significantly disrupt cell membranes, induce local inflammation, and impair autophagy processes, which collectively contribute to neuronal loss. As such, targeting AβOs specifically, rather than solely focusing on amyloid-β fibrils (AβFs), may offer a more effective therapeutic approach for AD. Recent advances in detection and diagnosis have emphasized the importance of accurately identifying AβOs in patient samples, enhancing the potential for timely intervention. In recent years, nanomaterials (NMs) have emerged as promising agents for addressing AβOs regarding their multivalent interactions, which can more effectively detect and inhibit AβO formation. This review provides an in-depth analysis of various nanochaperones developed to target AβOs, detailing their mechanisms of action and therapeutic potential via focusing on two main strategies, namely, disruption of AβOs through direct interaction and the inhibition of AβO nucleation by binding to intermediates of the oligomerization process. Evidence from in vivo studies indicate that NMs hold promise for ameliorating AD symptoms. Additionally, the review explores the different interaction mechanisms through which nanoparticles exhibit their inhibitory effects on AβOs, providing insights into their potential for clinical application. This comprehensive overview highlights the current advancements in NM-based therapies for AD and outlines future research directions aimed at optimizing these innovative treatments.
Collapse
Affiliation(s)
- Shiwani Randhawa
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Trilok Chand Saini
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Manik Bathla
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rahul Bhardwaj
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rubina Dhiman
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
| | - Amitabha Acharya
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
33
|
Papetti AV, Jin M, Ma Z, Stillitano AC, Jiang P. Chimeric brain models: Unlocking insights into human neural development, aging, diseases, and cell therapies. Neuron 2025:S0896-6273(25)00256-9. [PMID: 40300597 DOI: 10.1016/j.neuron.2025.03.036] [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: 01/26/2025] [Revised: 03/07/2025] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Human-rodent chimeric brain models serve as a unique platform for investigating the pathophysiology of human cells within a living brain environment. These models are established by transplanting human tissue- or human pluripotent stem cell (hPSC)-derived macroglial, microglial, or neuronal lineage cells, as well as cerebral organoids, into the brains of host animals. This approach has opened new avenues for exploring human brain development, disease mechanisms, and regenerative processes. Here, we highlight recent advancements in using chimeric models to study human neural development, aging, and disease. Additionally, we explore the potential applications of these models for studying human glial cell-replacement therapies, studying in vivo human glial-to-neuron reprogramming, and harnessing single-cell omics and advanced functional assays to uncover detailed insights into human neurobiology. Finally, we discuss strategies to enhance the precision and translational relevance of these models, expanding their impact in stem cell and neuroscience research.
Collapse
Affiliation(s)
- Ava V Papetti
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Alessandro C Stillitano
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA.
| |
Collapse
|
34
|
VandeBunte AM, Ortiz BL, Paolillo EW, Saloner R, Diaz V, Dutt S, Cadwallader CJ, Chen C, Lago AL, Rojas JC, Chan B, Sible I, Kramer JH, Casaletto KB. Relationships between blood pressure indicators and fluid biomarkers of brain aging in functionally intact older adults. Alzheimers Res Ther 2025; 17:85. [PMID: 40259431 PMCID: PMC12010523 DOI: 10.1186/s13195-025-01731-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/06/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Dementia risk is significantly shaped by cardiovascular health, with elevated blood pressure emerging as a key risk factor for adverse brain aging. Blood biomarkers such as pTau181, Aβ42/40, NfL, and GFAP have improved our understanding of dementia pathophysiology, however, few studies have explored how specific blood pressure metrics relate to biomarker levels, which could inform personalized dementia prevention strategies as these biomarkers move into clinic. We examined how different blood pressure metrics associated with molecular markers of astrocytic activation (GFAP), neuronal axon breakdown (NfL), and Alzheimer's disease pathobiology (pTau181, Aβ42/40) in plasma. METHODS 109 functionally intact (Clinical Dementia Rating Scale = 0) older adults completed blood draws with plasma assayed for Aβ42/40, GFAP, NfL, and pTau181 (Quanterix Simoa) and in-lab blood pressure quantification. Blood pressure metrics included diastolic blood pressure, systolic blood pressure, and pulse pressure (systolic minus diastolic). Separate regression models evaluated plasma biomarkers as a function of each blood pressure metric, adjusting for age and biological sex. Interaction models tested whether relationships between blood pressure metrics and plasma biomarkers differed by sex, age, or APOE-ε4 status. RESULTS With the exception of Aβ42/40, higher pulse pressure related to higher levels of all plasma biomarkers examined (pTau181, NfL, GFAP). Additionally, higher systolic blood pressure related to higher pTau181, while diastolic blood pressure did not meaningfully associate with any biomarker. Interaction models revealed a significantly stronger relationship between elevated pulse pressure and higher GFAP concentrations in females compared to males, as well as a significantly stronger association between elevated pulse pressure and lower Aβ42/40 plasma concentrations in APOE-ε4 carriers compared to non-carriers. CONCLUSIONS Our findings suggest that elevated pulse pressure, and to a lesser extent systolic blood pressure, are associated with increased Alzheimer's disease and neurodegenerative (axonal and astrocytic health) biology among typically aging adults. These associations underscore the importance of blood pressure management, particularly pulse pressure, for reducing dementia risk. Cardiovascular health may be incorporated with biomarkers to further personalize dementia prevention and management strategies.
Collapse
Affiliation(s)
- Anna M VandeBunte
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
- Palo Alto University, Palo Alto, CA, 94304, USA
| | - Bailey L Ortiz
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
- Palo Alto University, Palo Alto, CA, 94304, USA
| | - Emily W Paolillo
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Valentina Diaz
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Shubir Dutt
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Claire J Cadwallader
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Coty Chen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Argentina Lario Lago
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Brandon Chan
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Isabel Sible
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA
| | - Kaitlin B Casaletto
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, San Francisco, CA, 94158, USA.
| |
Collapse
|
35
|
Pang RK, Shi J, Peng XY, Su S, Zheng JY, Le K, Keng VW, Zhang SJ, Li XX. Huang-Lian-Jie-Du decoction alleviates cognitive deficits in Alzheimer's disease model 5xFAD mice by inhibiting Trem2/Dap12 signaling pathway. Chin Med 2025; 20:50. [PMID: 40234956 PMCID: PMC11998141 DOI: 10.1186/s13020-025-01098-x] [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: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder predominantly affecting the elderly population. It is characterized by cognitive deficits associated with the accumulation of amyloid-beta plaques and neurofibrillary tangles. Huang-Lian-Jie-Du (HLJD) decoction, recognized as a representative formulation with heat-clearing and detoxification effects, has been demonstrated to be effective in treating AD. However, the underlying mechanisms require further investigation. METHODS 5xFAD mice were administrated low and high doses of HLJD. The Morris water maze test was conducted to assess the effects of HLJD. Aβ42 and total tau protein levels were evaluated. Additionally, network pharmacology analysis was performed to identify therapeutic targets of HLJD's active components and their relevance to AD. ELISA, qPCR, Western Blot, and immunofluorescence assays were employed to confirm the identified pathways. Finally, primary microglia isolated from 5xFAD mice were used to validate the candidate targets of HLJD. RESULTS HLJD improved cognitive deficits in 5xFAD mice and reduced amyloid plaque deposition and tau protein levels. Network pharmacology analysis indicated that HLJD influences the neuroinflammatory response, particularly through the Dap12 signaling pathway. This was confirmed by reduced levels of neuroinflammation markers, including TNF-α, IL-1β, IL-6, and indicators of microglial activation and polarization. The expression of Trem2 and Dap12 in the hippocampus (HIP) of 5xFAD mice, as well as in the isolated primary microglia, were downregulated following HLJD treatment. CONCLUSION Our study indicates that HLJD alleviates cognitive deficits in AD by suppressing the Trem2/Dap12 signaling pathway in the HIP of 5xFAD mice, thereby inhibiting microglial neuroinflammation.
Collapse
Affiliation(s)
- Rui-Kang Pang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Jia Shi
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Xiang-Yu Peng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Shan Su
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Jia-Yi Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi, China
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hong Kong, SAR, China
| | - Vincent W Keng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China.
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China.
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China.
| | - Shi-Jie Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China.
| | - Xiao-Xiao Li
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China.
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China.
| |
Collapse
|
36
|
Ozsahin I, Wang X, Zhou L, Xi K, Hojjati SH, Tanzi E, Maloney T, Fung EK, Dyke JP, Chen K, Pahlajani S, McIntire LB, Costa AP, Dartora WJ, Razlighi QR, Glodzik L, Li Y, Chiang GC, Rusinek H, de Leon MJ, Butler TA. Divergent neurodegeneration associations with choroid plexus volume and degree of calcification in cognitively normal APOE ε4 carriers and non-carriers. Sci Rep 2025; 15:12818. [PMID: 40229453 PMCID: PMC11997051 DOI: 10.1038/s41598-025-97409-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/04/2025] [Indexed: 04/16/2025] Open
Abstract
Choroid plexus (CP), best known for producing CSF, also regulate inflammation and clear metabolic waste to maintain brain homeostasis. CP dysfunction is implicated in Alzheimer's Disease (AD), with MRI studies showing CP enlargement in AD. The basis for CP enlargement is unknown. We hypothesized that calcium deposition within CP, which increases with aging and in certain neurodegenerative conditions, might underlie pathologic CP enlargement and be linked to neurodegeneration. In 166 cognitively normal participants, we used multimodal imaging to examine CP structure (MRI-measured overall volume, CT-measured calcium volume), PET-measured Aβ, age, and APOE genotype as predictors of neurodegeneration, indexed as hippocampal volume. CP enlargement was associated with reduced hippocampal volume, particularly in APOE4 carriers. CP calcium was not independently associated with hippocampal volume. However, a significant interaction revealed APOE4 genotype-specific associations between CP calcium and neurodegeneration, with APOE4 carriers showing greater hippocampal volumes in association with greater CP calcium-opposite to our hypothesis. Results suggest that a factor other than calcium drives pathologic CP enlargement associated with neurodegeneration, with this factor especially important in APOE4 carriers. Candidate factors include lipids and inflammatory cells, which are known to accumulate in CP and be regulated by APOE. Our findings highlight CP as a critical locus for studying AD pathogenesis and the mechanisms by which APOE4 promotes AD.
Collapse
Affiliation(s)
- Ilker Ozsahin
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA.
- Operational Research Center in Healthcare, Near East University, Near East Boulevard, Nicosia/TRNC, 99138, Mersin 10, Turkey.
| | - Xiuyuan Wang
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Liangdong Zhou
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Ke Xi
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Seyed Hani Hojjati
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Emily Tanzi
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Thomas Maloney
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Edward K Fung
- Department of Radiology, Weill Cornell Medicine, 525 East 68th Street, New York, NY, 10065, USA
| | - Jonathan P Dyke
- Department of Radiology, Weill Cornell Medicine, 525 East 68th Street, New York, NY, 10065, USA
| | - Kewei Chen
- Banner Alzheimer Institute, Arizona State University, 901 E Willetta St, Phoenix, AZ, 85006, USA
| | - Silky Pahlajani
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Laura Beth McIntire
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Ana Paula Costa
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - William Jones Dartora
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Qolamreza R Razlighi
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Lidia Glodzik
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Yi Li
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Gloria C Chiang
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Henry Rusinek
- Department of Radiology, New York University, 660 1st Avenue, New York, NY, 10016, USA
| | - Mony J de Leon
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA
| | - Tracy A Butler
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, 407 East 61st Street, 2nd floor, New York, NY, 10065, USA.
| |
Collapse
|
37
|
Pappas I, Lohman T, Dutt S, Kapoor A, Engstrom AC, Alitin JPM, Barnes S, Chakhoyan A, Saca L, Gaggar R, Nourollahimoghadam E, Wang DJJ, Lai MHC, Joe EB, Ringman JM, Yassine HN, Schneider LS, Chui HC, Toga AW, Zlokovic BV, Nation DA. Cerebral hypoperfusion, brain structural integrity, and cognitive impairment in older APOE4 carriers. GeroScience 2025:10.1007/s11357-025-01642-5. [PMID: 40220152 DOI: 10.1007/s11357-025-01642-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Cerebral blood flow (CBF) deficits, cognitive decline, and brain structural changes have been reported in older adults with and without apolipoprotein E-e4 (APOE4)-related risk for dementia. However, it remains unclear whether brain structural changes mediate the effects of hypoperfusion on cognitive impairment in APOE4 carriers and non-carriers. We studied 166 (60-89 years) APOE4 carriers (ε3/ε4 or ε4/ε4) and APOE3 homozygotes (e3/e3) with and without cognitive impairment by clinical dementia rating (CDR) and neuropsychological testing. Pseudocontinuous arterial spin-labeling-MRI assessed regional CBF, and T1-anatomical and diffusion-MRI assessed structural integrity. Mediation analyses examined relationships among grey matter CBF, grey matter volume, and white matter integrity in regions underlying impairment in distinct cognitive ability domains. APOE4 carriers with global/memory impairment (CDR 0.5) exhibited decreased CBF in the posterior cingulate, decreased grey matter volume in the hippocampus, parahippocampal gyrus, and posterior cingulate, and decreased white matter integrity in the cingulum relative to APOE4 carriers with no impairment (CDR 0). Mediation analysis in APOE4 carriers indicated decreased posterior cingulate CBF effects on global/memory impairment were mediated by decreased cingulum integrity. In the combined APOE4 and APOE3 carriers sample, there were direct effects of frontal and inferior parietal CBF and superior longitudinal fasciculus integrity on attention/executive impairment. There were also direct effects of left inferior frontal CBF on language impairment. Findings suggest links between hypoperfusion and brain structural integrity underlying global/memory impairment in APOE4 carriers. Independent CBF relationships with structural integrity are also identified across genotypes and impairment domains.
Collapse
Affiliation(s)
- Ioannis Pappas
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Trevor Lohman
- Leonard Davis School of Gerontology, University of Southern California, Andrus Gerontology Center, 3715 McClintock Ave, Los Angeles, CA, 90089, USA
| | - Shubir Dutt
- Memory and Aging Center, University of California San Francisco, San Francisco, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Allison C Engstrom
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - John Paul M Alitin
- Leonard Davis School of Gerontology, University of Southern California, Andrus Gerontology Center, 3715 McClintock Ave, Los Angeles, CA, 90089, USA
| | - Samuel Barnes
- Department of Radiology, Loma Linda University, Loma Linda, CA, USA
| | - Ararat Chakhoyan
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lucas Saca
- Department of Radiology, Loma Linda University, Loma Linda, CA, USA
| | - Raghav Gaggar
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Elnaz Nourollahimoghadam
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Danny J J Wang
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mark H C Lai
- Deparment of Psychology, Dana and David Dornsife College of Arts and Letters, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth B Joe
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John M Ringman
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hussein N Yassine
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lon S Schneider
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - Helena C Chui
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel A Nation
- Leonard Davis School of Gerontology, University of Southern California, Andrus Gerontology Center, 3715 McClintock Ave, Los Angeles, CA, 90089, USA.
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
38
|
Gong K, Zhou S, Xiao L, Xu M, Zhou Y, Lu K, Yu X, Zhu J, Liu C, Zhu Q. Danggui Shaoyao San ameliorates Alzheimer's disease by regulating lipid metabolism and inhibiting neuronal ferroptosis through the AMPK/Sp1/ACSL4 signaling pathway. Front Pharmacol 2025; 16:1588375. [PMID: 40271063 PMCID: PMC12014676 DOI: 10.3389/fphar.2025.1588375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline; recent studies suggest that neuronal ferroptosis plays a key role in its pathogenesis. Danggui Shaoyao San (DSS), a traditional Chinese medicine formula, has shown demonstrated neuroprotective effects, but its precise mechanisms in AD treatment remain unclear. This study aims to investigate the mechanism of DSS in treating AD by inhibiting neuronal ferroptosis, explore whether DSS alleviates AD by suppressing neuronal ferroptosis via the AMPK/Sp1/ACSL4 pathway. Methods Chemical composition of DSS was identified by LC-MS/MS, followed by network pharmacology to predict targets and pathways. Molecular docking assessed binding affinities between DSS compounds and key proteins (AMPK, Sp1, ACSL4). In vivo experiments on APP/PS1 mice evaluated DSS effects on cognitive function, oxidative stress markers, lipid peroxidation, and ferroptosis-related proteins. Results Network pharmacology analysis suggested that DSS regulates lipid metabolism and inhibits neuronal ferroptosis via the AMPK pathway. Molecular docking revealed strong binding affinities between DSS compounds and AMPK downstream proteins, Sp1 and ACSL4. In vivo experiments showed that DSS improved cognitive function, enhanced antioxidant capacity, reduced lipid peroxide accumulation, and decreased Fe2+ content in brain tissue. Furthermore, DSS increased the expression of FTH, p-AMPK, and GPX4 while decreasing Sp1 and ACSL4 levels, thereby inhibiting ferroptosis. Conclusion DSS alleviates AD symptoms by suppressing neuronal ferroptosis via the AMPK/Sp1/ACSL4 axis, representing a novel lipid metabolism-targeted therapeutic strategy.
Collapse
Affiliation(s)
- Kai Gong
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuang Zhou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Li Xiao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengzhen Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuhe Zhou
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kaihui Lu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin Yu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiang Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Jinan, China
| |
Collapse
|
39
|
Li Z, Gong C. NLRP3 inflammasome in Alzheimer's disease: molecular mechanisms and emerging therapies. Front Immunol 2025; 16:1583886. [PMID: 40260242 PMCID: PMC12009708 DOI: 10.3389/fimmu.2025.1583886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory impairment, and neuroinflammation, with no definitive cure currently available. The NLRP3 inflammasome, a key mediator of neuroinflammation, has emerged as a critical player in AD pathogenesis, contributing to the accumulation of β-amyloid (Aβ) plaques, tau hyperphosphorylation, and neuronal damage. This review explores the mechanisms by which the NLRP3 inflammasome is activated in AD, including its interactions with Aβ, tau, reactive oxygen species (ROS), and pyroptosis. Additionally, it highlights the role of the ubiquitin system, ion channels, autophagy, and gut microbiota in regulating NLRP3 activation. Therapeutic strategies targeting the NLRP3 inflammasome, such as IL-1β inhibitors, natural compounds, and novel small molecules, are discussed as promising approaches to mitigate neuroinflammation and slow AD progression. This review underscores the potential of NLRP3 inflammasome inhibition as a therapeutic avenue for AD.
Collapse
Affiliation(s)
- Zhitao Li
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunrong Gong
- Department of Rehabilitation Medicine, Linyi People’s Hospital, Linyi, China
| |
Collapse
|
40
|
Huang Y, Xu J, Fan Z, Hu Y, He X, Chen A, Liu Y, Yin R, Guo J, DeKosky ST, Jaffee M, Zhou M, Su C, Wang F, Guo Y, Bian J. Identifying Alzheimer's Disease Progression Subphenotypes via a Graph-based Framework using Electronic Health Records. RESEARCH SQUARE 2025:rs.3.rs-6257332. [PMID: 40297697 PMCID: PMC12036456 DOI: 10.21203/rs.3.rs-6257332/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Purpose Understanding the heterogeneity of neurodegeneration in Alzheimer's disease (AD) development, as well as identifying AD progression pathways, is vital for enhancing diagnosis, treatment, prognosis, and prevention strategies. To identify disease progression subphenotypes in patients with mild cognitive impairment (MCI) and AD using electronic health records (EHRs). Methods We identified patients with mild cognitive impairment (MCI) and AD from the electronic health records from the OneFlorida+ Clinical Research Consortium. We proposed an outcome-oriented graph neural network-based model to identify progression pathways from MCI to AD. Results Of the included 2,525 patients, 61.66% were female, and the mean age was 76. In this cohort, 64.83% were Non-Hispanic White (NHW), 16.48% were Non-Hispanic Black (NHB), and 2.53% were of other races. Additionally, there were 274 Hispanic patients, accounting for 10.85% of the total patient population. The average duration from the first MCI diagnosis to the transition to AD was 891 days. We identified four progression subphenotypes, each with distinct characteristics. The average progression times from MCI to AD varied among these subphenotypes, ranging from 805 to 1,236 days. Conclusion The findings suggest that AD does not follow uniform transitions of disease states but rather exhibits heterogeneous progression pathways. Our proposed framework holds the potential to identify AD progression subphenotypes, providing valuable and explainable insights for the development of the disease.
Collapse
|
41
|
Cummings JL, Teunissen CE, Fiske BK, Le Ber I, Wildsmith KR, Schöll M, Dunn B, Scheltens P. Biomarker-guided decision making in clinical drug development for neurodegenerative disorders. Nat Rev Drug Discov 2025:10.1038/s41573-025-01165-w. [PMID: 40185982 DOI: 10.1038/s41573-025-01165-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 04/07/2025]
Abstract
Neurodegenerative disorders are characterized by complex neurobiological changes that are reflected in biomarker alterations detectable in blood, cerebrospinal fluid (CSF) and with brain imaging. As accessible proxies for processes that are difficult to measure, biomarkers are tools that hold increasingly important roles in drug development and clinical trial decision making. In the past few years, biomarkers have been the basis for accelerated approval of new therapies for Alzheimer disease and amyotrophic lateral sclerosis as surrogate end points reasonably likely to predict clinical benefit.Blood-based biomarkers are emerging for Alzheimer disease and other neurodegenerative disorders (for example, Parkinson disease, frontotemporal dementia), and some biomarkers may be informative across multiple disease states. Collection of CSF provides access to biomarkers not available in plasma, including markers of synaptic dysfunction and neuroinflammation. Molecular imaging is identifying an increasing array of targets, including amyloid plaques, neurofibrillary tangles, inflammation, mitochondrial dysfunction and synaptic density. In this Review, we consider how biomarkers can be implemented in clinical trials depending on their context of use, including providing information on disease risk and/or susceptibility, diagnosis, prognosis, pharmacodynamic outcomes, monitoring, prediction of response to therapy and safety. Informed choice of increasingly available biomarkers and rational deployment in clinical trials support drug development decision making and de-risk the drug development process for neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Brian K Fiske
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Göteborg, Sweden
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Billy Dunn
- The Michael J. Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Philip Scheltens
- Alzheimer's Center Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
- EQT Group, Dementia Fund, Stockholm, Sweden
| |
Collapse
|
42
|
Watts A, Donofry S, Ripperger H, Eklund NM, Wan L, Kang C, Grove G, Oberlin LE, Gujral S, Vidoni ED, Burns JM, McAuley E, Hillman CH, Kramer AF, Kamboh MI, Erickson KI. Lifetime estrogen exposure and domain-specific cognitive performance: results from the IGNITE study. Front Aging Neurosci 2025; 17:1524474. [PMID: 40248334 PMCID: PMC12003372 DOI: 10.3389/fnagi.2025.1524474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/06/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction Disruptions in estrogen exposure (i.e., surgically induced menopause) have been linked to poorer cognitive aging and dementia risk. Hormone therapy use (e.g., birth control, menopausal hormone therapy) has shown mixed associations with cognitive performance, possibly due to limited cognitive test batteries. To address previous inconsistencies, we investigated baseline data from Investigating Gains in Neurocognition in an Intervention Trial of Exercise (IGNITE). We hypothesized that (1) oophorectomy prior to natural menopause would be associated with poorer cognitive performance, (2) timing and duration of birth control and menopausal hormone therapy would influence associations with cognitive performance, and (3) APOE4 carrier status would interact with oophorectomy and hormone therapy to influence cognitive performance. Methods In 461 post-menopausal females (M age = 69.6) we assessed oophorectomy and hormone therapy use to examine associations with the Montreal Cognitive Assessment (MoCA) and factor-analytically derived composite scores for episodic memory, processing speed, working memory, executive function/attentional control, and visuospatial processing. Results Hypothesis (1) We did not observe associations between oophorectomy prior to natural menopause and poorer cognitive performance. However, hormone therapy use, started on average within 2 years of oophorectomy, was associated with better episodic memory (β = 0.106, p = 0.02), working memory (β = 0.120, p = 0.005), and visuospatial processing (β = 0.095, p = 0.03). Hypothesis (2) Birth control use was associated with better performance on the MoCA (β = 0.093, p = 0.04), working memory (β = 0.102, p = 0.02), and executive function/attentional control (β = 0.103, p = 0.02). However, duration and timing of birth control and menopausal hormone therapy were not associated with cognitive performance. Hypothesis (3) We did not observe significant interactions between APOE4 status and oophorectomy or hormone therapy in their associations with cognitive performance. Discussion Our results suggest exposure to estrogen during adulthood, specifically birth control and hormone therapy among women undergoing pre-menopausal oophorectomy, benefits cognitive function in older adulthood. Our comprehensive cognitive battery allowed us to examine cognitive function with a high degree of granularity. Future work should evaluate causal mechanisms of associations between lifetime estrogen exposure and later life cognitive function.
Collapse
Affiliation(s)
- Amber Watts
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Fairway, KS, United States
- Behavioral Research in Aging Neuroscience Cognition and Health (BRANCH) Lab, Psychology, University of Kansas, Lawrence, KS, United States
| | - Shannon Donofry
- Behavioral and Policy Sciences, Rand Corporation, Pittsburgh, PA, United States
- Brain Aging and Cognitive Health Lab, Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hayley Ripperger
- Brain Aging and Cognitive Health Lab, Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nicole M. Eklund
- Behavioral Neuroscience, Department of Graduate Medical Sciences, Boston University School of Medicine, Boston, MA, United States
| | - Lu Wan
- Neuroscience, Advent Health Research Institute, Orlando, FL, United States
| | - Chaeryon Kang
- Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - George Grove
- Brain Aging and Cognitive Health Lab, Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lauren E. Oberlin
- Neuroscience, Advent Health Research Institute, Orlando, FL, United States
- Psychiatry, Weill Cornell Medicine, New York, NY, United States
| | - Swathi Gujral
- Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Eric D. Vidoni
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Edward McAuley
- Health and Kinesiology, University of Illinois Urbana-Champaign, Urbana, IL, United States
- The Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Charles H. Hillman
- Center for Cognitive and Brain Health, Psychology, Northeastern University, Boston, MA, United States
| | - Arthur F. Kramer
- The Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Center for Cognitive and Brain Health, Psychology, Northeastern University, Boston, MA, United States
| | - M. Ilyas Kamboh
- Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kirk I. Erickson
- Brain Aging and Cognitive Health Lab, Psychology, University of Pittsburgh, Pittsburgh, PA, United States
- Neuroscience, Advent Health Research Institute, Orlando, FL, United States
| |
Collapse
|
43
|
Adil NA, Omo-Erigbe C, Yadav H, Jain S. The Oral-Gut Microbiome-Brain Axis in Cognition. Microorganisms 2025; 13:814. [PMID: 40284650 PMCID: PMC12029813 DOI: 10.3390/microorganisms13040814] [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: 01/16/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline and neuronal loss, affecting millions worldwide. Emerging evidence highlights the oral microbiome-a complex ecosystem of bacteria, fungi, viruses, and protozoa as a significant factor in cognitive health. Dysbiosis of the oral microbiome contributes to systemic inflammation, disrupts the blood-brain barrier, and promotes neuroinflammation, processes increasingly implicated in the pathogenesis of AD. This review examines the mechanisms linking oral microbiome dysbiosis to cognitive decline through the oral-brain and oral-gut-brain axis. These interconnected pathways enable bidirectional communication between the oral cavity, gut, and brain via neural, immune, and endocrine signaling. Oral pathogens, such as Porphyromonas gingivalis, along with virulence factors, including lipopolysaccharides (LPS) and gingipains, contribute to neuroinflammation, while metabolic byproducts, such as short-chain fatty acids (SCFAs) and peptidoglycans, further exacerbate systemic immune activation. Additionally, this review explores the influence of external factors, including diet, pH balance, medication use, smoking, alcohol consumption, and oral hygiene, on oral microbial diversity and stability, highlighting their role in shaping cognitive outcomes. The dynamic interplay between the oral and gut microbiomes reinforces the importance of microbial homeostasis in preserving systemic and neurological health. The interventions, including probiotics, prebiotics, and dietary modifications, offer promising strategies to support cognitive function and reduce the risk of neurodegenerative diseases, such as AD, by maintaining a diverse microbiome. Future longitudinal research is needed to identify the long-term impact of oral microbiome dysbiosis on cognition.
Collapse
Affiliation(s)
- Noorul Ain Adil
- USF Center for Microbiome Research, Microbiomes Institute, Tampa, FL 33612, USA; (N.A.A.); (C.O.-E.); (H.Y.)
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA
| | - Christabel Omo-Erigbe
- USF Center for Microbiome Research, Microbiomes Institute, Tampa, FL 33612, USA; (N.A.A.); (C.O.-E.); (H.Y.)
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, Tampa, FL 33612, USA; (N.A.A.); (C.O.-E.); (H.Y.)
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, Tampa, FL 33612, USA; (N.A.A.); (C.O.-E.); (H.Y.)
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
44
|
Hou X, Heckman MG, Fiesel FC, Koga S, Soto‐Beasley AI, Watzlawik JO, Zhao J, Valentino RR, Johnson PW, White LJ, Quicksall ZS, Reddy JS, Bras J, Guerreiro R, Zhao N, Bu G, Dickson DW, Ross OA, Springer W. Genome-wide association analysis identifies APOE as a mitophagy modifier in Lewy body disease. Alzheimers Dement 2025; 21:e70198. [PMID: 40309932 PMCID: PMC12044520 DOI: 10.1002/alz.70198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Phosphorylated ubiquitin (p-S65-Ub) is generated during PINK1-PRKN mitophagy as a specific marker of mitochondrial damage. Despite the widespread deposition of p-S65-Ub in aged and diseased human brain, the genetic contribution to its accumulation remains unclear. METHODS To identify novel mitophagy regulators, we performed a genome-wide association study using p-S65-Ub level as a quantitative trait in 1012 autopsy-confirmed Lewy body disease (LBD) samples. RESULTS We identified a significant genome-wide association with p-S65-Ub for rs429358 (apolipoprotein E ε4 [APOE4]) and a suggestive association for rs6480922 (ZMIZ1). APOE4 was associated with higher p-S65-Ub levels and greater neuropathological burden. Functional validation in mouse and human induced pluripotent stem cell (iPSC) models confirmed APOE4-mediated mitophagy alterations. Intriguingly, ZMIZ1 rs6480922 was associated with lower p-S65-Ub levels, reduced neuropathological load, and increased brain weight, indicating a potential protective role. DISCUSSION Our findings underscore the importance of mitochondrial quality control in LBD pathogenesis and nominate regulators that may contribute to disease risk or resilience. HIGHLIGHTS p-S65-Ub levels were used as a quantitative marker of mitochondrial damage. A GWAS identified two genetic variants that modify mitophagy in LBD autopsy brain. APOE4 was associated with increased p-S65-Ub accumulation and neuropathology. APOE4 altered mitophagy via pathology-dependent and pathology-independent mechanisms. ZMIZ1 was linked to reduced p-S65-Ub and neuropathology indicative of protection.
Collapse
Affiliation(s)
- Xu Hou
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Michael G. Heckman
- Division of Clinical Trials and BiostatisticsMayo ClinicJacksonvilleFloridaUSA
| | - Fabienne C. Fiesel
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Shunsuke Koga
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Jing Zhao
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Patrick W. Johnson
- Division of Clinical Trials and BiostatisticsMayo ClinicJacksonvilleFloridaUSA
| | - Launia J. White
- Division of Clinical Trials and BiostatisticsMayo ClinicJacksonvilleFloridaUSA
| | | | - Joseph S. Reddy
- Department of Quantitative Health SciencesMayo ClinicJacksonvilleFloridaUSA
| | - Jose Bras
- Department of Neurodegenerative ScienceVan Andel InstituteGrand RapidsMichiganUSA
- Division of Psychiatry and Behavioral MedicineMichigan State University College of Human MedicineGrand RapidsMichiganUSA
| | - Rita Guerreiro
- Department of Neurodegenerative ScienceVan Andel InstituteGrand RapidsMichiganUSA
- Division of Psychiatry and Behavioral MedicineMichigan State University College of Human MedicineGrand RapidsMichiganUSA
| | - Na Zhao
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Dennis W. Dickson
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Owen A. Ross
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| | - Wolfdieter Springer
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Neuroscience PhD ProgramMayo Clinic Graduate School of Biomedical SciencesJacksonvilleFloridaUSA
| |
Collapse
|
45
|
Lin YJ, Liu Y, Sheng ZH, Fu Y, Ma LZ, Zhang ZH, Wang LY, Huang LY, Liu M, Wang ZT, Tan L. The associations of cerebrospinal fluid ApoE and C1q with Alzheimer's disease biomarkers. J Alzheimers Dis 2025; 104:852-861. [PMID: 40091552 DOI: 10.1177/13872877251320419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
BackgroundThe roles of complement 1q (C1q) and Apolipoprotein E (ApoE) in driving Alzheimer's disease (AD) progression might be explained by their associations with neuroinflammation and AD pathology which were previously reported.ObjectiveWe examined the associations of cerebrospinal fluid (CSF) C1q and ApoE with CSF neuroinflammatory biomarkers and AD core biomarkers, as well as explored whether C1q mediated the associations of CSF ApoE with these biomarkers.MethodsHere, we analyzed CSF proteomics data from Alzheimer's Disease Neuroimaging Initiative (ADNI) using two different ADNI proteomics datasets-SomaScan (n = 579)and multiple reaction monitoring (MRM[n = 207]). Linear regression analyses were conducted to explore the association of CSF ApoE and C1q. The mediation model and structural equation model (SEM) were conducted to explore the associations of ApoE and C1q with AD biomarkers.ResultsMultiple linear regression showed that CSF ApoE was positively associated with CSF C1q in total participants and Alzheimer's continuum participants. Mediation analyses indicated that C1q mediated the associations of CSF ApoE with CSF T-tau, P-tau, sTREM2 and GFAP (mediation proportions range from 15.06 to 44.64%; all the p values < 0.05) but not with CSF amyloid-β and progranulin (PGRN). The SEM yielded similar results.ConclusionsOur findings suggest that C1q is linked to ApoE, and it mediates the associations of ApoE with T-tau, P-tau, sTREM2, GFAP, indicating C1q association with ApoE might be involved in AD progression.
Collapse
Affiliation(s)
- Yu-Jing Lin
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Ying Liu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ze-Hu Sheng
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ling-Zhi Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zi-Hao Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan-Yang Wang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Min Liu
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Dalian, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
46
|
Lonkar N, Latz E, McManus RM. Neuroinflammation and immunometabolism in neurodegenerative diseases. Curr Opin Neurol 2025; 38:163-171. [PMID: 39936491 DOI: 10.1097/wco.0000000000001356] [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: 02/13/2025]
Abstract
PURPOSE OF REVIEW Immunometabolism is an emerging field of research investigating the ability of immune cells to modulate their metabolic activity for optimal function. While this has been extensively examined in peripheral immune cells like macrophages, only recently have these studies been extended to assess the immunometabolic activity of microglia, the innate immune cells of the brain. RECENT FINDINGS Microglia are highly metabolically flexible and can utilize different nutrients for their diverse functions. Like other immune cells, they undergo metabolic reprogramming on immune stimulation and in inflammatory, neurodegenerative conditions such as Alzheimer's disease (AD). In recent years, researchers have looked at the intricate mechanisms that modulate microglial activity and have uncovered key links between altered metabolism, neuroinflammation, and the involvement of disease-associated risk genes. SUMMARY This review highlights the recent studies that have significantly contributed to our understanding of the metabolic dysregulation observed in activated microglia in conditions such as AD, unveiling novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Neha Lonkar
- German Center for Neurodegenerative Diseases (DZNE)
- Institute of Innate Immunity, University Hospital Bonn, Bonn
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, Bonn
- Deutsches Rheuma-Forschungszentrum (DRFZ), Berlin, Germany
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE)
- Institute of Innate Immunity, University Hospital Bonn, Bonn
| |
Collapse
|
47
|
Wu CT, Hu LH, Weng HY, Liu YM, Lin YF, Tsai SF, Lo RY, Ching YH. Rare APOE p.Gly4Glu: A putative disease-causing variant for early-onset Alzheimer's disease identified by next-generation sequencing. Tzu Chi Med J 2025; 37:175-180. [PMID: 40321968 PMCID: PMC12048122 DOI: 10.4103/tcmj.tcmj_117_24] [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: 05/08/2024] [Revised: 07/02/2024] [Accepted: 09/20/2024] [Indexed: 05/08/2025] Open
Abstract
Objectives We aimed to identify the early-onset Alzheimer's disease (EOAD)-causing variants in the Eastern Taiwanese population. Materials and Methods Twenty-one patients diagnosed with EOAD in the memory clinic at Hualien Tzu Chi Hospital were enrolled during 2014-2018. We conducted whole-exome sequencing to identify the disease-causing variations and validated by Sanger sequencing. SIFT, PolyPhen-2, and AlphaFold were applied to predict the functional impact of the identified variants. Results Two unrelated normolipidemic EOAD patients were carrying a rare heterozygous APOE variant (rs373985746, NC_000019.10:g. 44905879G>A, NM_001302688.2:c. 11G>A, and NP_001289617.1:p.Gly4Glu) with the allele frequency as 0.000206. Sanger sequencing uncovered the ∑ haplotypes in which the c.11G>A variation resided. SIFT predicted that the variant severely impacts protein structure and, maybe thus, function. AlphaFold predicted a dysfunctional conformation of the mutant APOE precursor a protein (p.Gly4Glu). Conclusion Our data strongly suggest that the rare p.Gly4Glu variant is associated with EOAD but does not cause dyslipidemia.
Collapse
Affiliation(s)
- Chu-Ting Wu
- Department of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Family Medicine, Puli Christian Hospital, Nantou, Taiwan
| | - Liang-Hsuan Hu
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Hui-Ying Weng
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Ming Liu
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Yung-Feng Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Shih-Feng Tsai
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli, Taiwan
| | - Raymond Y. Lo
- Department of Neurology, Taitung St. Mary’s Hospital, Taitung, Taiwan
- Department of Neurology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Biochemistry and Molecular Medicine, National Dong Hwa University, Hualien, Taiwan
| | - Yung-Hao Ching
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
48
|
Moseholm KF, Jensen MK, Buzkova P, Aroner SA, Fitzpatrick AL, Longstreth WT, Lopez O, Siscovick DS, Kizer JR, Ix JH, Hughes TM, Hayden KM, Nomura S, Tsai MY, McClelland R, Djoussé L, Mukamal KJ. Circulating non-esterified fatty acids, risk of dementia and cognitive decline: The cardiovascular health study and multi-ethnic study of atherosclerosis. Neurobiol Aging 2025; 148:71-79. [PMID: 39951847 DOI: 10.1016/j.neurobiolaging.2025.01.009] [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: 06/28/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025]
Abstract
Circulating non-esterified fatty acids (NEFAs) have toxic effects on a variety of organs central to cardiometabolic disease and can cross the blood-brain barrier. Whether NEFAs associate with cognitive decline or dementia remains unknown. Circulating total NEFA levels were measured in 3242 participants without dementia among older adults of the Cardiovascular Health Study (CHS) and related to adjudicated dementia over 6 years (n = 456 cases) and annually assessed cognitive decline. For confirmation, we related circulating NEFAs to cognition assessed 10 years later among 4361 participants in the Multi-Ethnic Study of Atherosclerosis (MESA). In CHS participants, each SD higher NEFA levels were associated with a hazard ratio (HR) for all-cause dementia of 1.11 (95 % CI: 1.01; 1.22). Baseline NEFA levels were also associated with more rapid decline in cognition over 6 years of follow-up. In MESA, circulating NEFA measurements were associated with lower cognitive scores measured 10 years later.'
Collapse
Affiliation(s)
- Kristine F Moseholm
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Denmark.
| | - Majken K Jensen
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Denmark; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Petra Buzkova
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Sarah A Aroner
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | - W T Longstreth
- Department of Epidemiology, University of Washington, Seattle, WA, USA; Department of Neurology, University of Washington, Seattle, WA, USA
| | - Oscar Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jorge R Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, and Departments of Medicine, Epidemiology, and Biostatistics, University of California, San Francisco, CA, USA
| | - Joachim H Ix
- Division of Nephrology and Hypertension, University of California, San Diego, CA, USA
| | - Timothy M Hughes
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kathleen M Hayden
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sarah Nomura
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Michael Y Tsai
- Division of Aging, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Robyn McClelland
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Luc Djoussé
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Kenneth J Mukamal
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Division of General Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
49
|
VandeBunte AM, Ortiz BL, Paolillo EW, Saloner R, Diaz V, Dutt S, Cadwallader CJ, Chen C, Lago AL, Rojas JC, Chan B, Sible I, Kramer JH, Casaletto KB. Relationships between blood pressure indicators and fluid biomarkers of brain aging in functionally intact older adults. RESEARCH SQUARE 2025:rs.3.rs-6018137. [PMID: 40196000 PMCID: PMC11975004 DOI: 10.21203/rs.3.rs-6018137/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Dementia risk is significantly shaped by cardiovascular health, with elevated blood pressure emerging as a key risk factor for adverse brain aging. Blood biomarkers such as pTau181, Aβ42/40, NfL, and GFAP have improved our understanding of dementia pathophysiology, however, few studies have explored how specific blood pressure metrics relate to biomarker levels, which could inform personalized dementia prevention strategies as these biomarkers move into clinic. We examined how different blood pressure metrics associated with molecular markers of astrocytic activation (GFAP), neuronal axon breakdown (NfL), and Alzheimer's disease pathobiology (pTau181, Aβ42/40) in plasma. Methods 109 functionally intact (Clinical Dementia Rating Scale=0) older adults completed blood draws with plasma assayed for Aβ42/40, GFAP, NfL, and pTau181 (Quanterix Simoa) and in-lab blood pressure quantification. Blood pressure metrics included diastolic blood pressure, systolic blood pressure, and pulse pressure (systolic minus diastolic). Separate regression models evaluated plasma biomarkers as a function of each blood pressure metric, adjusting for age and biological sex. Interaction models tested whether relationships between blood pressure metrics and plasma biomarkers differed by sex, age, or APOE-ε4 status. Results With the exception of Aβ42/40, higher pulse pressure related to higher levels of all plasma biomarkers examined (pTau181, NfL, GFAP). Additionally, higher systolic blood pressure related to higher pTau181, while diastolic blood pressure did not meaningfully associate with any biomarker. Interaction models revealed a significantly stronger relationship between elevated pulse pressure and higher GFAP concentrations in females compared to males, as well as a significantly stronger association between elevated pulse pressure and lower Aβ42/40 plasma concentrations in APOE-ε4 carriers compared to non-carriers. Conclusions Our findings suggest that elevated pulse pressure, and to a lesser extent systolic blood pressure, are associated with increased Alzheimer's disease and neurodegenerative (axonal and astrocytic health) biology among typically aging adults. These associations underscore the importance of blood pressure management, particularly pulse pressure, for reducing dementia risk. Cardiovascular health may be incorporated with biomarkers to further personalize dementia prevention and management strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Coty Chen
- University of California, San Francisco
| | | | | | | | | | | | | |
Collapse
|
50
|
Li Y, Izhar T, Kanekiyo T. HDAC3 as an Emerging Therapeutic Target for Alzheimer's Disease and other Neurological Disorders. Mol Neurobiol 2025:10.1007/s12035-025-04866-w. [PMID: 40126601 DOI: 10.1007/s12035-025-04866-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the aged population. Histone acetylation is a major epigenetic mechanism linked to memory formation and cognitive function. Histone deacetylases (HDACs) are responsible for the deacetylation of lysine residues in histone proteins. Although pan-HDAC inhibitors are effective in ameliorating AD phenotypes in preclinical models, they are associated with potential unfavorable adverse effects and barely translated into clinical trials. Therefore, the development of novel HDAC inhibitors with a well isoform-selectivity has been desired in AD drug discovery. Among various HDAC isoforms, HDAC3 is highly expressed in neurons and exhibits detrimental effects on synaptic plasticity and cognitive function. Moreover, HDAC3 provokes neuroinflammation and neurotoxicity and contributes to AD pathogenesis. In this review, we highlight HDAC3 as an attractive therapeutic target for disease-modifying therapy in AD. In addition, we discuss the therapeutic potential of HDAC3 inhibitors in other neurological disorders.
Collapse
Affiliation(s)
- Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Taha Izhar
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| |
Collapse
|