1
|
Xie J, Van Hoecke L, Van Wonterghem E, Van Imschoot G, Andries V, Vereecke L, Vandenbroucke RE. The gut-brain axis in Alzheimer's disease is shaped by commensal gut microbiota derived extracellular vesicles. Gut Microbes 2025; 17:2501193. [PMID: 40355382 PMCID: PMC12077474 DOI: 10.1080/19490976.2025.2501193] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/31/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025] Open
Abstract
Emerging clinical and experimental evidence highlight the involvement of gut microbiota in the onset and progression of neurodegenerative diseases such as Alzheimer's disease (AD) via neuroinflammatory processes along the gut-brain axis. Despite this, the precise mechanisms governing gut microbial involvement in AD remain elusive. In this study, we observed that AppNL-G-F AD mice raised under germ-free (GF) conditions, display a reduced amyloid-β (Aβ) pathology, accompanied by a shift in microglial cells toward a less inflammatory state and increased phagocytotic efficiency. In addition, we demonstrate that gut microbiota depletion can protect against synaptic deficits in AD mice. Notably, administering bacterial extracellular vesicles (bEVs), i.e. nano-sized particles packed with bacterial components, derived from fecal slurry from specific pathogen-free housed AppNL-G-F AD mice, reversed the effects of GF conditions on both microglial activation and Aβ plaque accumulation. These findings reveal for the first time that commensal gut microbiota-derived bEVs have a major impact on AD pathology progression.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Griet Van Imschoot
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Vanessa Andries
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent University, Ghent, Belgium
| | - Lars Vereecke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent University, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent University, Ghent, Belgium
| |
Collapse
|
2
|
Jin Q, Wang S, Yao Y, Jiang Q, Li K. The gut-eye axis: from brain neurodegenerative diseases to age-related macular degeneration. Neural Regen Res 2025; 20:2741-2757. [PMID: 39435619 PMCID: PMC11826455 DOI: 10.4103/nrr.nrr-d-24-00531] [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: 05/09/2024] [Revised: 07/25/2024] [Accepted: 09/21/2024] [Indexed: 10/23/2024] Open
Abstract
Age-related macular degeneration is a serious neurodegenerative disease of the retina that significantly impacts vision. Unfortunately, the specific pathogenesis remains unclear, and effective early treatment options are consequently lacking. The microbiome is defined as a large ecosystem of microorganisms living within and coexisting with a host. The intestinal microbiome undergoes dynamic changes owing to age, diet, genetics, and other factors. Such dysregulation of the intestinal flora can disrupt the microecological balance, resulting in immunological and metabolic dysfunction in the host, and affecting the development of many diseases. In recent decades, significant evidence has indicated that the intestinal flora also influences systems outside of the digestive tract, including the brain. Indeed, several studies have demonstrated the critical role of the gut-brain axis in the development of brain neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Similarly, the role of the "gut-eye axis" has been confirmed to play a role in the pathogenesis of many ocular disorders. Moreover, age-related macular degeneration and many brain neurodegenerative diseases have been shown to share several risk factors and to exhibit comparable etiologies. As such, the intestinal flora may play an important role in age-related macular degeneration. Given the above context, the present review aims to clarify the gut-brain and gut-eye connections, assess the effect of intestinal flora and metabolites on age-related macular degeneration, and identify potential diagnostic markers and therapeutic strategies. Currently, direct research on the role of intestinal flora in age-related macular degeneration is still relatively limited, while studies focusing solely on intestinal flora are insufficient to fully elucidate its functional role in age-related macular degeneration. Organ-on-a-chip technology has shown promise in clarifying the gut-eye interactions, while integrating analysis of the intestinal flora with research on metabolites through metabolomics and other techniques is crucial for understanding their potential mechanisms.
Collapse
Affiliation(s)
- Qianzi Jin
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Suyu Wang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yujia Yao
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qin Jiang
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Keran Li
- Department of Ophthalmology, The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
3
|
Machado AM, Ferraz AS, Pereira MG, Leite F. Beyond the beats: a systematic review of the underlying inflammatory pathways between atrial fibrillation and cognitive decline. Neurol Sci 2025; 46:2951-2963. [PMID: 39976882 DOI: 10.1007/s10072-025-08040-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/31/2025] [Indexed: 06/11/2025]
Abstract
Atrial fibrillation (AF) and cognitive decline represent significant health challenges with increasing prevalence and significant socioeconomic implications. Emerging evidence suggests a potential link between AF and cognitive decline, including dementia and Alzheimer's disease, although the underlying mechanisms remain incompletely understood. Inflammation has emerged as a key mediator in cardiovascular and neurological diseases, encouraging an investigation into its role in the atrial fibrillation-cognition association. A systematic search of PubMed, Web of Science, and PsycInfo was conducted to identify relevant studies investigating possible inflammatory mechanisms bridging AF and cognitive decline. Studies were assessed for quality and relevance, and data were synthesized using a narrative approach. Five papers were included, with only two longitudinal studies. Inflammatory biomarkers emerged as significant factors associated with both AF and cognitive decline. Three studies revealed a correlation between high-sensitivity CRP (HS-CRP) levels and cognitive decline in patients with AF, AF patients with cerebral infarction, and elderly individuals with AF. However, conflicting results were observed, as one study did not identify any associations between cognitive decline and HS-CRP levels. The scientific literature on this topic is scarce, and the results of existing studies often lack consistency in their findings, highlighting the need for further research to better understand and prevent this significant health burden in patients with AF. So, the results of this study are expected to inform future research directions and cognitive decline risk stratification, guiding the development of targeted interventions aimed at preserving cognitive function and improving outcomes in patients with AF.
Collapse
Affiliation(s)
- Ana Mónica Machado
- Research Centre in Psychology (Cipsi), School of Psychology, University of Minho, Braga, Portugal
| | - Ana Sofia Ferraz
- Research Centre in Psychology (Cipsi), School of Psychology, University of Minho, Braga, Portugal
| | - M Graça Pereira
- Research Centre in Psychology (Cipsi), School of Psychology, University of Minho, Braga, Portugal
| | - Fernanda Leite
- Department of Transfusion Medicine, Santo António University Hospital Center, Porto, Portugal.
- Public Health and Forensic Sciences, and Medical Education Department, Faculty of Medicine University of Porto, Porto, Portugal.
- i3S-Institute for Research and Innovation in Health, Porto, Portugal.
| |
Collapse
|
4
|
Renesteen E, Boyajian JL, Islam P, Kassab A, Abosalha A, Makhlouf S, Santos M, Chen H, Shum-Tim C, Prakash S. Microbiome Engineering for Biotherapeutic in Alzheimer's Disease Through the Gut-Brain Axis: Potentials and Limitations. Int J Mol Sci 2025; 26:5351. [PMID: 40508160 PMCID: PMC12154405 DOI: 10.3390/ijms26115351] [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: 04/15/2025] [Revised: 05/21/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition characterized by considerable cognitive decline and functional impairment, primarily due to the progressive alteration of neurons, microglia, and astrocytes. Pathological manifestations of AD include the loss of synaptic plasticity, reduction in synaptic strength by amyloid-beta, aggregation, and neurotoxicity from tau protein post-translational modifications, all contributing to the disruption of neural networks. Despite its current pharmacological treatment for AD, different approaches to treat such disease are being developed, from a microbiome perspective. The microbiome encompasses a diverse microorganism, including beneficial bacteria that create a positive impact to diminish AD pathogenesis. Growing evidence suggests that probiotic, prebiotic, synbiotic, and postbiotics can positively modulate the gut-brain axis, reducing systemic inflammation, restoring neurotransmitter balance, and improving gut health, thereby possibly mitigating AD pathogenesis. Moreover, there is paraprobiotics as the most recently developed biotherapeutic with beneficial effects. This review explores the correlation between AD and gut-brain axis as a novel biotherapeutic target. The underlying mechanism of the microbiota-gut-brain axis in AD is examined. Novel insights into the current applications as potential treatment and its limitations are highlighted.
Collapse
Affiliation(s)
- Editha Renesteen
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Jacqueline L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Ahmed Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta Al-Geish St., The Medical Campus, Tanta 31527, Egypt
| | - Stephanie Makhlouf
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Madison Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Hongmei Chen
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Cedrique Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 2B4, Canada; (E.R.); (J.L.B.); (A.K.); (S.M.)
| |
Collapse
|
5
|
Kazemi M, Esmaeili-Mahani S, Abbasnejad M, Sheibani V. Neurotrophic factor neuritin ameliorates streptozotocin-induced Alzheimer's disease-like impairment of memory, neuroinflammation, apoptotic factors and compensates hippocampal neuritin expression. Behav Brain Res 2025; 486:115542. [PMID: 40127821 DOI: 10.1016/j.bbr.2025.115542] [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: 08/24/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/26/2025]
Abstract
Alzheimer's disease (AD) is the main cause of dementia in the elderly, and is becoming one of the most expensive and deadly diseases. Deficiency of neurotrophic factors signaling is an important cause of this disease. Therefore, we investigated whether neuritin as a neurotrophic factor can have a neuroprotective effect against streptozotocin (STZ)-induced rat model of AD. The animals were bilaterally injected with intra hippocampal-STZ (3 mg/kg). Different concentrations of neuritin (0.5, 1, 1.5 µg/rat) were administrated 15 min before STZ injection. After 14 days, the rats were evaluated for cognitive performance using novel object recognition (NOR), open field and Morris water maze (MWM) tests and then sacrificed for biochemical analysis (by real-time PCR and western blot examinations). The results demonstrated that the STZ- induced learning and memory impairments were significantly prevented by 1.5 µg neuritin. Moreover, the increased levels of inflammatory factors (NF-κb, TNF-α and IL-1β) and apoptotic parameters (cytochrome c and caspase‑3) in STZ- treated rats were also significantly decreased by neuritin. In addition, hippocampal neuritin gene expression was downregulated by STZ injection, which was reversed by intra hippocampal neuritin injection. In conclusion, the present study suggests that neuritin prevents cognitive defects in AD rat model and its expression level is associated with cognitive resilience.
Collapse
Affiliation(s)
- Mandana Kazemi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Saeed Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran; Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehdi Abbasnejad
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Vahid Sheibani
- Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
6
|
Fołta J, Rzepka Z, Wrześniok D. The Role of Inflammation in Neurodegenerative Diseases: Parkinson's Disease, Alzheimer's Disease, and Multiple Sclerosis. Int J Mol Sci 2025; 26:5177. [PMID: 40507988 PMCID: PMC12154089 DOI: 10.3390/ijms26115177] [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: 04/17/2025] [Revised: 05/15/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025] Open
Abstract
Neurodegenerative diseases are a group of conditions that have in common the progressive damage and degeneration of neurons in the central nervous system. This group includes Parkinson's disease, Alzheimer's disease, and multiple sclerosis, among others. In recent years, increasing evidence has pointed to the important role of inflammation in the pathogenesis of these conditions. The occurrence of inflammation in the brain, which is often triggered by pro-inflammatory activation of microglia or astrocytes, can consequently lead to a chronic inflammatory response that contributes to the development of neurodegenerative processes. Inflammatory processes themselves, both within the nervous system and throughout the human body, appear to be central to the initiation and progression of neuronal damage. Understanding the role of inflammation in these diseases may open up new perspectives and opportunities in the future in the development of effective therapies to improve patients' quality of life as the vast majority of cases of patients affected by neurodegenerative diseases continue to be treated symptomatically since causal treatments are lacking. In this review, we provide information on the impact of inflammation on the pathogenesis, course, and potential therapeutic options for selected neurodegenerative diseases. In addition, this article provides a general description of neuroinflammation and the involvement and role of specific cells in the central nervous system.
Collapse
Affiliation(s)
| | | | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland; (J.F.); (Z.R.)
| |
Collapse
|
7
|
Yang W, Yu Q, Wang N, Lam KK, Lin ZX, Xian YF. Far-Infrared Radiation Ameliorates the Cognitive Dysfunction in an Alzheimer's Disease Transgenic Mouse via Modulating Jak-2/Stat3 and Nrf-2/HO-1 Pathways. Neuromolecular Med 2025; 27:34. [PMID: 40374872 DOI: 10.1007/s12017-025-08860-2] [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: 04/02/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
Alzheimer's disease (AD) is the primary cause of dementia in the elderly. However, effective therapies that modify the disease process in AD remain elusive. Far-infrared radiation (FIR) is commonly utilized as a complementary treatment a range of disease, for example insomnia and rheumatoid arthritis. In this research, we explored how FIR light impacts the cognitive functions of TgCRND8 AD mice and elucidated its underlying molecular mechanism. The cognitive capabilities of TgCRND8 mice assessed by employing the Morris water maze. The concentrations of IL-1β, TNF-α, IL-4, Aβ40, and Aβ42 protein were assessed by enzyme-linked immunosorbent assay. Immunostaining was conducted to assess the Aβ deposits and microglial presence in the brains of TgCRND8 mice. Western blot was applied to detect the protein expressions of tau phosphorylation, amyloid-β (Aβ) production, Jak-2/Stat3, and Nrf-2/HO-1 pathways. The results indicated that FIR light notably ameliorated the cognitive impairments of the AD mice, reduced both Aβ deposition and tau protein hyperphosphorylation at sites of Thr205, Ser369, Ser404, and Thr181, suppressed the release of TNF-α and IL-1β, attenuated the ratios of p-Jak-2/Jak-2 and p-Stat3/Stat3, while increased the protein levels of IL-4, Nrf-2, and HO-1 in the brains of TgCRND8 mice. These findings amply demonstrated that FIR light ameliorated cognitive deficits of TgCRND8 mice via reducing both Aβ burden and tau protein hyperphosphorylation, suppressing the neuroinflammation, and restoring the levels of the oxidative-related proteins through modulating Jak-2/Stat3 and Nrf-2/HO-1 pathways. These experimental findings indicate that FIR light treatment is a promising treatment approach for AD.
Collapse
Affiliation(s)
- Wen Yang
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Qiuxia Yu
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Nick Wang
- Nick Wang Technology Limited, TML Tower, 3 Hoi Shing Road, Tsuen Wan, Kowloon, Hong Kong SAR, People's Republic of China
| | - Koon Kit Lam
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People's Republic of China.
| | - Yan-Fang Xian
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| |
Collapse
|
8
|
Xu R, Gao Y. CD28 + CD45RA - CD8br AC mediated the effects of Interleukin- 6 on Alzheimer's disease: A Mendelian Randomization Study. BMC Neurol 2025; 25:203. [PMID: 40369481 PMCID: PMC12077015 DOI: 10.1186/s12883-025-04194-5] [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: 05/06/2024] [Accepted: 04/14/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND IL-6 has garnered significant attention as a potential factor in AD pathogenesis. The association between peripheral immune cells and IL-6 is evident, yet how peripheral immune cells mediate IL-6's effects on AD remains enigmatic regarding the precise pathophysiological processes. To address these uncertainties, we employed genetic evidence to investigate their impact. Our current study explores further the intricate relationship between IL-6, peripheral immune cells, and AD by using extensive publicly available genetic data, aiming to provide novel insights into this critical area of medical research. METHODS The relevant data regarding IL-6, 731 peripheral immune cells, and AD were screened and retrieved from the GWAS database. Subsequently, we predominantly utilized the IVW approach to carry out bi-directional MR analyses between IL-6, 731 peripheral immune cells, and AD. We utilized two-step, two-sample MR analyses to determine three key factors: (i) IL-6 exhibits associations with both AD and specific peripheral immune cells, respectively, and there is an absence of inverse causality. (ii) Specific peripheral immune cells exhibit associations with AD, and there is an absence of inverse causality. (iii) to identify which peripheral immune cells mediate the effects of IL-6 on AD. Then we employed the MVMR approach to verify whether the mediating relationships obtained from the two-step, two-sample MR analyses remained valid. Furthermore, we calculated their respective mediating effects, the combined mediating effects, and the proportions of their mediating effect shares. All of the aforementioned steps were utilized to verify the reliability of causality employing sensitivity analysis, heterogeneity analysis, and horizontal pleiotropy analysis. RESULTS Our findings indicate a significant correlation between increased IL-6 levels and a reduced risk of AD (P = 0.009, OR = 0.941, 95%CI = 0.899- 0.985), along with elevated levels of CD28+ CD45RA- CD8br AC (P = 0.007, OR = 1.159, 95%CI = 1.007- 1.333). Also indicates a significant correlation between increased CD28+ CD45RA- CD8br AC (P = 0.005, OR = 0.983, 95%CI = 0.971- 0.995) levels and a reduced risk of AD. Therefore, through MVMR analysis, the effect of IL-6 on AD increased from -0.061 to -0.046 (95% CI: -0.090, -0.002) after genetic adjustment for CD28+ CD45RA- CD8br AC. CONCLUSIONS Increased CD28+ CD45RA- CD8br AC levels appear to partially mediate the effect of IL-6 on reducing AD risk.
Collapse
Affiliation(s)
- Rui Xu
- Chengdu Sixth People's Hospital, Chengdu City, Sichuan Province, China.
| | - YongJun Gao
- The Second Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, China
| |
Collapse
|
9
|
Yordanov Y, Tzankova V, Stefanova D, Georgieva M, Tzankova D. Exploratory Data Analysis of the In Vitro Effects of Novel Hydrazide-Hydrazone Antioxidants in the Context of In Silico Predictors. Antioxidants (Basel) 2025; 14:566. [PMID: 40427448 PMCID: PMC12108285 DOI: 10.3390/antiox14050566] [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: 04/04/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Substantial in vitro experimental data have been produced about the safety, antioxidant, neuro- and hepatoprotective effects of a series of recently synthesized N-pyrrolyl hydrazide-hydrazones (compounds 5, 5a-5g). However, compound activity across multiple assays varies and it is challenging to elucidate the favorable physicochemical characteristics of the studied compounds and guide further lead optimization. The aim of the current study is to apply exploratory data analysis in order to profile the biological effects of the novel hydrazide-hydrazones, gain insights related to their mechanisms of action in the context of in silico predictions and identify key predictor-outcome relationships. We collected a dataset from available in vitro studies of compounds 5, 5a-5g. It included cytotoxicity values, protection against hydrogen peroxide-induced damage in HepG2 and SH-SY5Y cells, two radical scavenging assays and a hemolysis assay across a range of treatment concentrations. SwissADME-based predictions of chemometric and ADME parameters and pro-oxidant enzyme docking data were generated to provide context for the interpretation of in vitro outcome patterns and identify causal relationships. Multiple factor analysis (MFA), followed by hierarchical clustering on principal components (HCPC), was applied to profile compounds' biological behavior. This revealed that differences in the number of H-bond donors, in the permeability coefficient and in the docking scores to two pro-oxidant enzymes could aid in explaining the effects of compounds with similar in vitro profiles. HCPC differentiated 5a as mostly neuroprotective, 5 and 5d as hepatoprotective radical scavengers, 5g with higher docking affinity to 5-lipoxygenase (5-LOX) and myeloperoxidase (MPO) and 5b, 5c and 5f as having less H-bond donors and variable in vitro activity. The consensus application of three variable selection approaches based on standard lasso regression, robust penalized regression and random forest confirmed the relationships between some in vitro outcomes and LogP, pan-assay interference (PAINS) alerts, 5-LOX allosteric site docking and H-bond donor numbers. The exploratory analysis of the combined in vitro and in silico dataset provides useful insights which could help explain the major drivers behind the experimental results. It can be informative in the design of new, improved members of the series of novel N-pyrrolyl hydrazide-hydrazones with better neuroprotective potential and less side effects.
Collapse
Affiliation(s)
- Yordan Yordanov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University—Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria (D.S.)
| | - Virginia Tzankova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University—Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria (D.S.)
| | - Denitsa Stefanova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University—Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria (D.S.)
| | - Maya Georgieva
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University—Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
| | - Diana Tzankova
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University—Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria
| |
Collapse
|
10
|
Koriath CA, Perneczky R. Obesity and Alzheimer's disease dementia: Examining inflammatory links to cognitive decline and neuropsychiatric symptoms. J Alzheimers Dis 2025:13872877251338467. [PMID: 40329584 DOI: 10.1177/13872877251338467] [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: 05/08/2025]
Abstract
Obesity is recognized as a risk factor for cardiovascular disease, vascular dementia, and Alzheimer's disease dementia (AD dementia). Emerging evidence indicates that obesity in AD patients is associated with heightened neuropsychiatric symptoms, as reflected by inflammatory biomarkers such as C-reactive protein and complement C3. Neuroinflammation, particularly through certain aspects of microglial activation, plays a significant role in AD development and cognitive decline. While further research is warranted to explore these neuroinflammatory pathways as potential therapeutic targets, proactive weight management starting in middle age may help mitigate both cognitive decline and neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Carolin Am Koriath
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
11
|
Yang B, Teymur A, Tang C, Wu T. V-set and immunoglobulin domain containing 4 as a potential predictor of Alzheimer's disease and advanced aging. J Alzheimers Dis 2025; 105:453-463. [PMID: 40116682 DOI: 10.1177/13872877251329463] [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: 03/23/2025]
Abstract
BackgroundV-set and immunoglobulin domain containing 4 (VSIG4) emerges as a significant player in the immune system pathways. It has been previously identified as a potential hub gene for Alzheimer's disease (AD) and aging, underscoring its importance in understanding these conditions.ObjectiveThis study aimed to evaluate the diagnostic potential of serum VSIG4 and identify trends in serum VSIG4 in relationship with other biomarkers and neurological tests.MethodsELISA was used to measure the serum concentration of VSIG4 in AD, compared to healthy subjects. The relationship between VSIG4 levels and the age of the subjects, as well as other AD-related serum proteins and various measures of cognition was examined.ResultsVSIG4 was significantly elevated in the serum of AD patients compared to healthy controls (p = 0.0074). Significant correlations were identified between serum VSIG4 and other notable proteins related to AD and inflammation, such as total tau, neurofilament light (NfL), YKL-40, CD14, FABP3, and TNF-α. Significant correlations were also identified between VSIG4 concentration and the results of neurological tests.ConclusionsSerum VSIG4 may reflect neuroinflammation and altered lipid processing, affecting the cognitive performance of AD and aging.
Collapse
Affiliation(s)
- Bowen Yang
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Aygun Teymur
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Chenling Tang
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| |
Collapse
|
12
|
Bashirrohelleh MA, Bavarsad K, Khodadadi A, Shohan M, Asadirad A. Curcumin-enhanced stem cell exosomes: A novel approach to modulating neuroinflammation and improving cognitive function in a rat model of Alzheimer's disease. Eur J Pharmacol 2025; 999:177695. [PMID: 40315951 DOI: 10.1016/j.ejphar.2025.177695] [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: 11/25/2024] [Revised: 04/22/2025] [Accepted: 04/30/2025] [Indexed: 05/04/2025]
Abstract
The effect of Curcumin-enhanced stem cell exosomes on the learning and memory impairment induced by streptozotocin (STZ) and neuro-inflammation in rats was evaluated. An animal model of Alzheimer's disease (AD) was established by intracerebroventricular (ICV) injection of STZ (3 mg/kg) in male Wistar rats (250 ± 50 g). ICV STZ injections chronically reduce cerebral glucose uptake and produce other effects similar to pathological, molecular and behavioral features of AD. Numerous studies confirmed the anti-inflammatory and antioxidant properties of curcumin (a natural polyphenol) against free radicals, as well as its ability to inhibit the aggregation of proteins such as beta-amyloid and alpha-synuclein in disorders such as AD and Parkinson's disease. The use of extracellular vesicles has garnered a lot of interest in research studies because of the important roles that mesenchymal stem cell-derived exosomes play in permeability, retention, and drug delivery as well as their ability to reduce inflammatory cytokines (TNF-α, IL-1β, and IL-6). Furthermore, researches highlighted the positive effect of curcumin on neuronal differentiation of stem cells in vivo and in vitro. Since studies emphasized the ameliorating effect of curcumin-treated macrophage-exosomes on symptoms of Alzheimer's disease by inhibiting tau protein phosphorylation, we proposed that Curcumin-primed MSC exosomes may offer greater efficacy to alleviate AD compared to naïve MSC exosomes. In this study, we investigated the effect of curcumin in stimulating the anti-inflammatory potential of exosome-derived stem cells. We evaluated the effect of MSC-EXO and pre-treated MSC-EXO with curcumin (CUR-MSC-EXO) on inhibiting inflammation and memory and learning impairments. Following four intraperitoneal injections of MSC-EXO and CUR-MSC-EXO at a dosage of 30μg/body over 30 days, we found that MSC-EXO and CUR-MSC-EXO elevated anti-inflammatory cytokines (IL10, TGF-β) and reduced pro-inflammatory cytokines (IL1, TNF-α) in peripheral blood compared to the AD group. The elevated level of M2 anti-inflammatory microglia markers (Arg1, CD206) and decreased level expression of M1 pro-inflammatory markers (iNOS, CD86) indicated that the CUR-MSC-EXO effect was more significant in the polarization of microglia into the M2 phenotype in the rat hippocampus. Both treatment groups demonstrated improvements in memory and learning skills. The results of the passive avoidance learning in the rats with STZ-induced memory impairment, however, were better in the CUR-MSC-EXO. Additionally, after therapy, a decrease in degenerative neurons was seen. Therefore, using curcumin may stimulate the anti-inflammatory and neuroprotective potential of exosome-derived stem cells which could provide hope for Alzheimer's disease treatment in the future.
Collapse
Affiliation(s)
- Mohammad-Ali Bashirrohelleh
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur, University of Medical Sciences, Iran; Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Kowsar Bavarsad
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Persian Gulf Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Khodadadi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur, University of Medical Sciences, Iran; Cancer, Petroleum and Environmental Pollutants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Shohan
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur, University of Medical Sciences, Iran
| | - Ali Asadirad
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur, University of Medical Sciences, Iran; Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Cancer, Petroleum and Environmental Pollutants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
13
|
Volloch V, Rits-Volloch S. Alzheimer's Is a Multiform Disease of Sustained Neuronal Integrated Stress Response Driven by the C99 Fragment Generated Independently of AβPP; Proteolytic Production of Aβ Is Suppressed in AD-Affected Neurons: Evolution of a Theory. Int J Mol Sci 2025; 26:4252. [PMID: 40362488 PMCID: PMC12073115 DOI: 10.3390/ijms26094252] [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: 03/18/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The present Perspective analyzes the remarkable evolution of the Amyloid Cascade Hypothesis 2.0 (ACH2.0) theory of Alzheimer's disease (AD) since its inception a few years ago, as reflected in the diminishing role of amyloid-beta (Aβ) in the disease. In the initial iteration of the ACH2.0, Aβ-protein-precursor (AβPP)-derived intraneuronal Aβ (iAβ), accumulated to neuronal integrated stress response (ISR)-eliciting levels, triggers AD. The neuronal ISR, in turn, activates the AβPP-independent production of its C99 fragment that is processed into iAβ, which drives the disease. The second iteration of the ACH2.0 stemmed from the realization that AD is, in fact, a disease of the sustained neuronal ISR. It introduced two categories of AD-conventional and unconventional-differing mainly in the manner of their causation. The former is caused by the neuronal ISR triggered by AβPP-derived iAβ, whereas in the latter, the neuronal ISR is elicited by stressors distinct from AβPP-derived iAβ and arising from brain trauma, viral and bacterial infections, and various types of inflammation. Moreover, conventional AD always contains an unconventional component, and in both forms, the disease is driven by iAβ generated independently of AβPP. In its third, the current, iteration, the ACH2.0 posits that proteolytic production of Aβ is suppressed in AD-affected neurons and that the disease is driven by C99 generated independently of AβPP. Suppression of Aβ production in AD seems an oxymoron: Aβ is equated with AD, and the later is inconceivable without the former in an ingrained Amyloid Cascade Hypothesis (ACH)-based notion. But suppression of Aβ production in AD-affected neurons is where the logic leads, and to follow it we only need to overcome the inertia of the preexisting assumptions. Moreover, not only is the generation of Aβ suppressed, so is the production of all components of the AβPP proteolytic pathway. This assertion is not a quantum leap (unless overcoming the inertia counts as such): the global cellular protein synthesis is severely suppressed under the neuronal ISR conditions, and there is no reason for constituents of the AβPP proteolytic pathway to be exempted, and they, apparently, are not, as indicated by the empirical data. In contrast, tau protein translation persists in AD-affected neurons under ISR conditions because the human tau mRNA contains an internal ribosomal entry site in its 5'UTR. In current mouse models, iAβ derived from AβPP expressed exogenously from human transgenes elicits the neuronal ISR and thus suppresses its own production. Its levels cannot principally reach AD pathology-causing levels regardless of the number of transgenes or the types of FAD mutations that they (or additional transgenes) carry. Since the AβPP-independent C99 production pathway is inoperative in mice, the current transgenic models have no potential for developing the full spectrum of AD pathology. What they display are only effects of the AβPP-derived iAβ-elicited neuronal ISR. The paper describes strategies to construct adequate transgenic AD models. It also details the utilization of human neuronal cells as the only adequate model system currently available for conventional and unconventional AD. The final alteration of the ACH2.0, introduced in the present Perspective, is that AβPP, which supports neuronal functionality and viability, is, after all, potentially produced in AD-affected neurons, albeit not conventionally but in an ISR-driven and -compatible process. Thus, the present narrative begins with the "omnipotent" Aβ capable of both triggering and driving the disease and ends up with this peptide largely dislodged from its pedestal and retaining its central role in triggering the disease in only one, although prevalent (conventional), category of AD (and driving it in none). Among interesting inferences of the present Perspective is the determination that "sporadic AD" is not sporadic at all ("non-familial" would be a much better designation). The term has fatalistic connotations, implying that the disease can strike at random. This is patently not the case: The conventional disease affects a distinct subpopulation, and the basis for unconventional AD is well understood. Another conclusion is that, unless prevented, the occurrence of conventional AD is inevitable given a sufficiently long lifespan. This Perspective also defines therapeutic directions not to be taken as well as auspicious ways forward. The former category includes ACH-based drugs (those interfering with the proteolytic production of Aβ and/or depleting extracellular Aβ). They are legitimate (albeit inefficient) preventive agents for conventional AD. There is, however, a proverbial snowball's chance in hell of them being effective in symptomatic AD, lecanemab, donanemab, and any other "…mab" or "…stat" notwithstanding. They comprise Aβ-specific antibodies, inhibitors of beta- and gamma-secretase, and modulators of the latter. In the latter category, among ways to go are the following: (1) Depletion of iAβ, which, if sufficiently "deep", opens up a tantalizing possibility of once-in-a-lifetime preventive transient treatment for conventional AD and aging-associated cognitive decline, AACD. (2) Composite therapy comprising the degradation of C99/iAβ and concurrent inhibition of the neuronal ISR. A single transient treatment could be sufficient to arrest the progression of conventional AD and prevent its recurrence for life. Multiple recurrent treatments would achieve the same outcome in unconventional AD. Alternatively, the sustained reduction/removal of unconventional neuronal ISR-eliciting stressors through the elimination of their source would convert unconventional AD into conventional one, preventable/treatable by a single transient administration of the composite C99/iAβ depletion/ISR suppression therapy. Efficient and suitable ISR inhibitors are available, and it is explicitly clear where to look for C99/iAβ-specific targeted degradation agents-activators of BACE1 and, especially, BACE2. Directly acting C99/iAβ-specific degradation agents such as proteolysis-targeting chimeras (PROTACs) and molecular-glue degraders (MGDs) are also viable options. (3) A circumscribed shift (either upstream or downstream) of the position of transcription start site (TSS) of the human AβPP gene, or, alternatively, a gene editing-mediated excision or replacement of a small, defined segment of its portion encoding 5'-untranslated region of AβPP mRNA; targeting AβPP RNA with anti-antisense oligonucleotides is another possibility. If properly executed, these RNA-based strategies would not interfere with the protein-coding potential of AβPP mRNA, and each would be capable of both preventing and stopping the AβPP-independent generation of C99 and thus of either preventing AD or arresting the progression of the disease in its conventional and unconventional forms. The paper is interspersed with "validation" sections: every conceptually significant notion is either validated by the existing data or an experimental procedure validating it is proposed.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
14
|
Wang S, Ni J, Wei M, Li T, Shi J, Tian J. Impact of obesity on neuropsychiatric symptoms in Alzheimer's disease: Insights from the ADNI cohort. J Alzheimers Dis 2025:13872877251331974. [PMID: 40261292 DOI: 10.1177/13872877251331974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
BackgroundObesity is a major global health issue linked to increased risks of dementia, including Alzheimer's disease (AD). While the association between obesity and neuropsychiatric symptoms (NPS) in AD remains underexplored, identifying these links could aid in weight management in AD patients.ObjectiveThis study investigates the relationship between body mass index (BMI) and NPS in AD dementia patients, focusing on the potential mediating role of systemic inflammation.MethodsWe employed Generalized Additive Models (GAMs) to explore the relationship between BMI and NPS, as measured by the Neuropsychiatric Inventory Questionnaire (NPI-Q). Participants were classified into ideal, overweight, and obese groups based on WHO criteria. Longitudinal analyses assessed the trajectory of NPI-Q scores in different groups over a one-year follow-up.ResultsBMI significantly affects NPI-Q total scores and specific symptoms, including delusions, hallucinations, agitation/aggression, elation/euphoria, disinhibition, irritability/lability, aberrant motor behavior, nighttime disturbances, and appetite/eating disturbances. Obese patients exhibited higher NPI-Q total scores and greater severity in symptoms such as hallucinations, agitation/aggression, elation/euphoria, apathy/indifference, disinhibition, aberrant motor behavior, and nighttime disturbances. Additionally, CRP and complement C3 were identified as mediators in the relationship between obesity and NPS, highlighting the role of systemic inflammation.ConclusionsThis study demonstrates that obesity is associated with a heightened burden of NPS in AD dementia patients. The identification of CRP and complement C3 as mediators suggests inflammation plays a crucial role in the association between obesity and NPS. These findings underscore the importance of addressing obesity and its inflammatory consequences in managing NPS among this vulnerable population.
Collapse
Affiliation(s)
- Shuoshi Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingnian Ni
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingqing Wei
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Li
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Shi
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinzhou Tian
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Kelekar U, Pan X, Philippou E, Muhammad T, Heyn PC, Wu B. Associations between oral health measures and cognitive function in middle aged and older Indian adults. Sci Rep 2025; 15:13183. [PMID: 40240374 PMCID: PMC12003837 DOI: 10.1038/s41598-025-90756-z] [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: 06/21/2024] [Accepted: 02/14/2025] [Indexed: 04/18/2025] Open
Abstract
Given India's rise in ageing population and emerging epidemiological evidence of the possible association between poor oral health and cognitive impairment, the present study examined associations between oral health measures and cognitive function using a cross-sectional dataset from Longitudinal Aging Study in India, 2017-2018. Analysis included 56,738 participants aged ≥ 45 years old. Self-reported physician-diagnosed oral health measures included loss of teeth (some or all), presence of painful teeth, ulcers, bleeding and swelling gums, loose teeth, dental cavities or caries, and soreness or cracks in the corners of the mouth. Cognitive function was assessed using a battery of neurocognitive tests across five domains: memory, orientation, arithmetic function, executive function, and object naming. Findings from multivariable regression models revealed that participants who lost all their teeth had lower cognition scores by 0.65 points [β= -0.65, 95% CI: -0.97, -0.33] than those who had not lost any teeth. An unexpected positive association was found between three or more oral conditions and cognitive performance [β = 0.53, 95% CI: 0.28, 0.77] that needs further investigation. Findings suggest that oral health is associated with cognitive function and there is a need to promote policies targeted at improving the oral health infrastructure and health education.
Collapse
Affiliation(s)
- Uma Kelekar
- School of Business, College of Business, Innovation, Leadership and Technology, Marymount University, Arlington, VA, USA.
- Center for Optimal Aging, Marymount University, Arlington, VA , United States.
| | - Xi Pan
- Department of Sociology, Texas State University, San Marcos, TX, 78666, USA
| | - Elena Philippou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - T Muhammad
- Department of Human Development and Family Studies , Center for Healthy Aging, Pennsylvania State University, , University Park PA 16802, United States
| | - Patricia C Heyn
- Center for Optimal Aging, Marymount University, Arlington, VA , United States
| | - Bei Wu
- Rory Meyers College of Nursing, New York University, New York, United States
| |
Collapse
|
16
|
Morisaki M, Rezaali F, Lau LC, Boche D, Khandaker GM, Turecki G, Sinclair LI. Depression as a risk factor for Alzheimer's disease: A human post-mortem study. PLoS One 2025; 20:e0320561. [PMID: 40179065 PMCID: PMC11967934 DOI: 10.1371/journal.pone.0320561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Depression is associated with persistent low mood. In mid to late life, it has been identified as a risk factor for Alzheimer's disease (AD) with evidence that depression might be an early manifestation of AD. Although the underlying mechanisms by which depression enhances AD development remain unknown, there are several features commonly seen in both diseases such as the presence of neuroinflammation. In this study, we aimed to identify whether neuroinflammation is increased in depression as observed in the early stages of AD by examining post-mortem human brain tissue. Post-mortem human brain tissue from 54 cases with depression and 37 controls without depression were retrieved from the Douglas Bell Canada Brain Bank. Sixteen early-stage AD cases defined as a Braak stage III-IV and 15 controls were sourced from the South West Dementia Brain Bank. Frozen tissue from the dorsal prefrontal cortex was obtained for all cases in order to measure inflammatory proteins (IFN-γ, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, and TNF-α) and endothelial markers (ICAM-1, VCAM) using ELISA and MesoScale Multiplex Assays. In the depression group, increase of IL-6 and IL-10, and decrease of IL-1β were observed compared to controls, with no changes detected for the other cytokines and the endothelial markers. In early-stage AD cases, only increased ICAM-1 expression was found compared to controls, indicating endothelial activation as an early feature of AD. None of the cytokines measured showed alteration of their expression in early-stage AD cases. Depression, but not AD, was associated with evidence of neuroinflammation. Depression may increase AD risk through different mechanism(s) than inflammation.
Collapse
Affiliation(s)
- Mizuki Morisaki
- Dementia Research Group, University of Bristol, Bristol, United Kingdom
| | - Farnoosh Rezaali
- Dementia Research Group, University of Bristol, Bristol, United Kingdom
| | - Laurie C. Lau
- Clinical and Experimental Sciences, Faculty of Medicine, Sir Henry Wellcome Laboratories, University of Southampton, Southampton, United Kingdom
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Golam M. Khandaker
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
- NIHR Bristol Biomedical Research Centre, Bristol, United Kingdom
| | - Gustavo Turecki
- Douglas Bell Brain Bank, McGill University, Montreal, Canada
| | | |
Collapse
|
17
|
Pesce G, Gusto G, Johansen P, Khachatryan A, Lopez-Ledesma B, Vukmirica J, Cases A. Systemic inflammation prevalence in patients with atherosclerotic cardiovascular disease and chronic kidney disease: a population-based study using a nationwide primary care database in Spain. Front Cardiovasc Med 2025; 12:1538466. [PMID: 40104140 PMCID: PMC11913838 DOI: 10.3389/fcvm.2025.1538466] [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: 12/02/2024] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
Introduction Systemic inflammation is recognised as a critical driver of atherosclerotic cardiovascular disease (ASCVD), especially in patients with comorbid chronic kidney disease (CKD). This study aims to assess the prevalence of systemic inflammation in the ASCVD population in Spain. Methods Outpatient electronic medical records from The Health Improvement Network (THIN®) database were used to identify patients with ASCVD and a C-reactive protein (CRP) measurement ≥1 between January 2014 and July 2023 in Spain. The proportion of patients with systemic inflammation (defined as CRP ≥ 2 mg/L) was estimated at the first CRP measurement (index date) and at the end of the study. The patients' characteristics, comorbidities, and drug dispensation in the prior 12 months were reported by systemic inflammation status at the index date. Results Overall, 15,798 patients with ASCVD were included in the study (mean age: 71.1 years; 57% men), of whom 34% had CKD. The proportion of patients with systemic inflammation at the index date was 58% (65% among CKD patients) and 56% (62% among CKD patients) at the end of the study. Patients with systemic inflammation were more frequently smokers, obese, with comorbidities, and had higher low-density lipoprotein cholesterol and triglycerides levels than patients without systemic inflammation. Overall, patients with ASCVD and systemic inflammation used statins and aspirin less frequently compared to patients without systemic inflammation, while they used antibiotics, anticoagulants, and antihypertensives more frequently. Conclusion Systemic inflammation prevalence is high among patients with ASCVD in Spain, especially among patients with comorbid CKD. Therapeutic strategies focused on targeting systemic inflammation may have beneficial effects in reducing the burden of ASCVD.
Collapse
Affiliation(s)
- Giancarlo Pesce
- Real-World Evidence & Modeling Solutions, Certara, Milan, Italy
| | - Gaelle Gusto
- Real-World Evidence & Modeling Solutions, Certara, Paris, France
| | | | - Artak Khachatryan
- Real-World Evidence & Modeling Solutions, Certara, London, United Kingdom
| | | | | | - Aleix Cases
- Servicio de Nefrologia, Hospital Clínic, Barcelona, Spain
| |
Collapse
|
18
|
Barrett-Young A, Reuben A, Caspi A, Cheyne K, Ireland D, Kokaua J, Ramrakha S, Tham YC, Theodore R, Wilson G, Wong TY, Moffitt T. Measures of retinal health successfully capture risk for Alzheimer's disease and related dementias at midlife. J Alzheimers Dis 2025:13872877251321114. [PMID: 40033783 DOI: 10.1177/13872877251321114] [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] [Indexed: 03/05/2025]
Abstract
BACKGROUND Identification of at-risk individuals who would benefit from early intervention for Alzheimer's disease and related dementias (ADRD) is critical as new treatments are developed. Measures of retinal health could offer accessible and low-cost indication of pre-morbid disease risk, but their association with ADRD risk is unknown. OBJECTIVE To determine whether midlife retinal neuronal and microvascular measures are associated with ADRD risk-index scores and individual domains of ADRD risk. METHODS Data were from the Dunedin Multidisciplinary Health and Development Study, a population-representative longitudinal New Zealand-based birth cohort study. 94.1% (N = 938) of living Study members were seen at age 45 (2017-2019). Retinal neuronal (retinal nerve fiber layer (RNFL) and ganglion cell-inner plexiform layer (GC-IPL)) and microvascular (arterioles and venules) measures were used as predictors. Outcome measures were four top ADRD risk indexes (CAIDE, LIBRA, Lancet, and ADU-ADRI), and a comprehensive midlife ADRD risk index, the DunedinARB. RESULTS Poorer retinal microvascular health (narrower arterioles and wider venules) was associated with greater ADRD risk (βs = 0.16-0.31; ps < 0.001). Thinner RNFL was modestly associated with higher ADRD risk (βs = 0.05-0.08; ps = 0.02-0.13). Follow-up tests of distinct domains of ADRD risk indicated that while RNFL associations reflected cardiometabolic risk only, microvascular measures were associated with diverse ADRD risk factors. CONCLUSIONS Measures of retinal health, particularly microvascular measures, successfully capture ADRD risk across several domains of known risk factors, even at the young midlife age of 45 years. Retinal microvascular imaging may be an accessible, scalable, and relatively low-cost method of assessing ADRD risk among middle-aged adults.
Collapse
Affiliation(s)
| | - Aaron Reuben
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Avshalom Caspi
- Department of Psychology & Neuroscience, Duke University, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
- PROMENTA, Department of Psychology, University of Oslo, Norway
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Kirsten Cheyne
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - David Ireland
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Jesse Kokaua
- Va'a o Tautai-Centre for Pacific Health, University of Otago, Dunedin, New Zealand
| | - Sandhya Ramrakha
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Yih-Chung Tham
- Singapore National Eye Centre, Singapore Eye Research Institute, Singapore
- Centre for Innovation and Precision Eye Health, Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Ophthalmology and Visual Science Academic Clinical Program, Duke-NUS Medical School, Singapore
| | | | - Graham Wilson
- Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Tien Yin Wong
- Singapore National Eye Centre, Singapore Eye Research Institute, Singapore
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Terrie Moffitt
- Department of Psychology & Neuroscience, Duke University, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, King's College London, London, UK
- PROMENTA, Department of Psychology, University of Oslo, Norway
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| |
Collapse
|
19
|
Blicharz-Futera K, Kamiński M, Grychowska K, Canale V, Zajdel P. Current development in sulfonamide derivatives to enable CNS-drug discovery. Bioorg Chem 2025; 156:108076. [PMID: 39889550 DOI: 10.1016/j.bioorg.2024.108076] [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: 10/06/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 02/03/2025]
Abstract
The encouraging therapeutic potential of sulfonamide-based derivatives has been unraveled by breakthrough discovery of Paul Ehrlich, who pointed out the possibility of fighting microbes with chemicals. Over the decades, the utility of sulfonamides has expanded beyond antimicrobial agents, revealing their usefulness in many areas of pharmacotherapy, including the treatment of central nervous system (CNS) diseases. Through a detailed analysis of preclinical and clinical data, we identify key sulfonamide-based compounds that have demonstrated significant CNS activity. We also discuss the challenges in the development of sulfonamide derivatives as enzyme/ion channel inhibitors or receptor ligands for CNS applications, describing their mode of action and therapeutic significance. This is followed by the characteristics of pharmacological targets, structure-activity relationships, ADMET properties, efficacy in experimental animal models, and outcomes from clinical trials. Overall, the versatile nature of arylsulfonamides makes them a valuable motif in drug discovery, offering diverse opportunities for the development of novel agents for treating CNS disorders.
Collapse
Affiliation(s)
- Klaudia Blicharz-Futera
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Łazarza Street, 31-530 Krakow, Poland
| | - Michał Kamiński
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Łazarza Street, 31-530 Krakow, Poland
| | - Katarzyna Grychowska
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Vittorio Canale
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Paweł Zajdel
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland.
| |
Collapse
|
20
|
Meng T, Zhang Y, Ye Y, Li H, He Y. Bioinformatics insights into mitochondrial and immune gene regulation in Alzheimer's disease. Eur J Med Res 2025; 30:89. [PMID: 39920860 PMCID: PMC11806906 DOI: 10.1186/s40001-025-02297-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND There is growing evidence that the pathogenesis of Alzheimer's disease is closely linked to the resident innate immune cells of the central nervous system, including microglia and astrocytes. Mitochondrial dysfunction in microglia has also been reported to play an essential role in the pathogenesis of AD and other neurological diseases. Therefore, finding the mitochondrial and immune-related gene (MIRG) signatures in AD can be significant in diagnosing and treating AD. METHODS In this study, the intersection of the differentially expressed genes (DEGs) from the GSE109887 cohort, immune-related genes (IRGs) obtained from WGCNA analysis, and mitochondria-related genes (MRGs) was taken to identify mitochondria-immune-related genes (MIRGs). Then, using machine learning algorithms, biomarkers with good diagnostic value were selected, and a nomogram was constructed. Subsequently, we further analyzed the signaling pathways and potential biological mechanisms of the biomarkers through gene set enrichment analysis, prediction of transcription factors (TFs), miRNAs, and drug prediction. RESULTS Using machine learning algorithms, five biomarkers (TSPO, HIGD1A, NDUFAB1, NT5DC3, and MRPS30) were successfully identified, and a nomogram model with strong diagnostic ability and accuracy (AUC > 0.9) was constructed. In addition, single-gene enrichment analysis revealed that NDUFAB1 was significantly enriched in pathways associated with diseases, such as Alzheimer's and Parkinson's, providing valuable insights for future clinical research on Alzheimer's in the context of mitochondrial-immune interactions. Interestingly, brain tissue pathology showed neuronal atrophy and demyelination in AD mice, along with a reduction in Nissl bodies. Furthermore, the escape latency of AD mice was significantly longer than that of the control group. After platform removal, there was a notable increase in the path complexity and time required to reach the target quadrant, suggesting a reduction in spatial memory capacity in AD mice. Moreover, qRT-PCR validation confirmed that the mRNA expression of the five biomarkers was consistent with bioinformatics results. In AD mice, TSPO expression was increased, while HIGD1A, NDUFAB1, NT5DC3, and MRPS30 expressions were decreased. However, peripheral blood samples did not show expression of HIGD1A or MRPS30. These findings provide new insights for research on Alzheimer's disease in the context of mitochondrial-immune interactions, further exploring the pathogenesis of Alzheimer's disease and offering new perspectives for the clinical development of novel drugs. CONCLUSIONS Five mitochondrial and immune biomarkers, i.e., TSPO, HIGD1A, NDUFAB1, NT5DC3, and MRPS30, with diagnostic value in Alzheimer's disease, were screened by machine-learning algorithmic models, which will be a guide for future clinical research of Alzheimer's disease in the mitochondria-immunity-related direction.
Collapse
Affiliation(s)
- Tian Meng
- Yunnan Yunke Institute of Biotechnology, No. 871 Longquan Rd, Kunming, 650500, China
| | - Yazhou Zhang
- Department of Geriatrics, The Second People's Hospital of Kunming, No. 338Guangming Rd, Kunming, 650233, Yunnan, China
| | - Yuan Ye
- Department of Geriatrics, The Second People's Hospital of Kunming, No. 338Guangming Rd, Kunming, 650233, Yunnan, China
| | - Hui Li
- Yunnan Labreal Biotechnology Co., LTD, No. 871 Longquan Rd, Kunming, 650500, China
| | - Yongsheng He
- Yunnan Yunke Institute of Biotechnology, No. 871 Longquan Rd, Kunming, 650500, China.
- Yunnan Labreal Biotechnology Co., LTD, No. 871 Longquan Rd, Kunming, 650500, China.
| |
Collapse
|
21
|
Sun M, Li F, Wang Y, Miao M, Lu Z, Chen WM, Wu SY, Zhang J. Postoperative sepsis and its sequential impact on dementia. Crit Care 2025; 29:66. [PMID: 39915868 PMCID: PMC11800527 DOI: 10.1186/s13054-025-05276-z] [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: 12/04/2024] [Accepted: 01/14/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Postoperative sepsis is a severe complication associated with increased mortality and potential long-term cognitive decline, including dementia. However, the relationship between postoperative sepsis and dementia remains poorly understood. METHODS This retrospective cohort study used data from the National Database in Taiwan, covering the period from January 1, 2005, to December 31, 2022. The index period for surgeries was set between January 1, 2008, and December 31, 2013, allowing the identification of patients without prior dementia. A landmark period of 12 months following surgery was defined to capture the number of postoperative sepsis events, which were then analyzed for their impact on dementia risk. After 1:4 propensity score matching (PSM), dementia and mortality were evaluated using Cox proportional hazards and Fine-Gray competing risk models. RESULTS Following PSM, 778 patients were in the postoperative sepsis group and 3,112 in the non-postoperative sepsis group. Dementia incidence was higher in the postoperative sepsis group (26%) compared to the non- postoperative sepsis group (13.6%), with a hazard ratio (HR) of 1.25 (95% CI, 1.03-1.52). A dose-response relationship was observed, with dementia rates of 24.5% for one postoperative sepsis event and 34.9% for two or more events, the latter showing an HR of 1.77 (95% CI, 1.17-2.66). Mortality was also elevated in the postoperative sepsis group (40.5% vs. 31.6%; HR 1.45, 95% CI, 1.28-1.65). CONCLUSIONS Postoperative sepsis is significantly associated with increased dementia risk in a dose-dependent manner. These findings highlight the importance of enhancing perioperative infection control to reduce both immediate and long-term cognitive complications.
Collapse
Affiliation(s)
- Mingyang Sun
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 7 Weiwu Rd, Zhengzhou, Henan, China
| | - Fangfang Li
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 7 Weiwu Rd, Zhengzhou, Henan, China
| | - Yangyang Wang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 7 Weiwu Rd, Zhengzhou, Henan, China
| | - Mengrong Miao
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 7 Weiwu Rd, Zhengzhou, Henan, China
| | - Zhongyuan Lu
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 7 Weiwu Rd, Zhengzhou, Henan, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan, China
| | - Wan-Ming Chen
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan
- Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan
| | - Szu-Yuan Wu
- Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, No. 83, Nanchang St., Luodong Township, Yilan County, 265, Taiwan.
- Division of Radiation Oncology, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan.
- Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan.
- Cancer Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan.
- Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan.
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 7 Weiwu Rd, Zhengzhou, Henan, China.
| |
Collapse
|
22
|
Cai H, Zhao T, Pang Y, Fu X, Ren Z, Quan S, Jia L. Systemic inflammatory markers in ageing, Alzheimer's disease and other dementias. Brain 2025; 148:480-492. [PMID: 39008616 DOI: 10.1093/brain/awae230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 05/28/2024] [Accepted: 06/23/2024] [Indexed: 07/17/2024] Open
Abstract
Systemic inflammation with alterations in inflammatory markers is involved in ageing and Alzheimer's disease. However, few studies have investigated the longitudinal trajectories of systemic inflammatory markers during ageing and Alzheimer's disease, and specific markers contributing to Alzheimer's disease remain undetermined. In this study, a longitudinal cohort (cohort 1: n = 290; controls, 136; preclinical Alzheimer's disease, 154) and a cross-sectional cohort (cohort 2: n = 351; controls, 62; Alzheimer's disease, 63; vascular dementia, 58; Parkinson's disease dementia, 56; behavioural variant frontotemporal dementia, 57; dementia with Lewy bodies, 55) were included. Plasma levels of inflammatory markers were measured every 2 years during a 10-year follow-up in the longitudinal cohort and once in the cross-sectional cohort. The study demonstrated that the inflammatory markers significantly altered during both ageing and the development of Alzheimer's disease. However, only complement C3, interleukin-1β and interleukin-6 exhibited significant changes in participants with preclinical Alzheimer's disease, and their longitudinal changes were significantly associated with the development of Alzheimer's disease compared to controls over the 10-year follow-up. In the cross-sectional cohort, complement C3 demonstrated specificity to Alzheimer's disease, while interleukin-1β and interleukin-6 were also altered in other dementias. The study provides a new perspective on the involvement of inflammatory markers in the ageing process and the development of Alzheimer's disease, implying that regulating inflammation may have a pivotal role in promoting successful ageing and in the prevention and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Huimin Cai
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Tan Zhao
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Yana Pang
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Xiaofeng Fu
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Ziye Ren
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Shuiyue Quan
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Longfei Jia
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| |
Collapse
|
23
|
Fang B, Wang Z, Nan G. Dietary inflammatory potential and the risk of cognitive impairment: A meta-analysis of prospective cohort studies. J Nutr Health Aging 2025; 29:100428. [PMID: 39689376 DOI: 10.1016/j.jnha.2024.100428] [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: 09/11/2024] [Revised: 11/02/2024] [Accepted: 11/22/2024] [Indexed: 12/19/2024]
Abstract
OBJECTIVE Dietary inflammatory potential, measured by the dietary inflammatory index (DII) has been linked to cognitive impairment. However, evidence was mostly driven by cross-sectional studies. This meta-analysis of prospective cohort studies aims to evaluate the relationship between DII and the risk of cognitive impairment, including mild cognitive impairment (MCI) and dementia. METHODS We conducted a systematic search of PubMed, Web of Science, and Embase for studies published up to July 25, 2024. Prospective cohort studies with adults aged 18 years or older, without dementia at baseline, and reporting the incidence of cognitive impairment by DII category were included. Data were analyzed using a random-effects model to calculate pooled risk ratios (RRs) with 95% confidence intervals (CIs). RESULTS Nine prospective cohort studies with 266,169 participants were included. A high DII at baseline was associated with an increased risk of cognitive impairment during follow-up (RR: 1.34, 95% CI: 1.15-1.55, p < 0.001) with moderate heterogeneity (I² = 56%). Subgroup analyses revealed consistent associations across types of cognitive impairment (MCI, overall dementia, Alzheimer's disease) and study characteristics (p for subgroup difference all >0.05). Sensitivity analyses confirmed the robustness of the results. CONCLUSIONS This meta-analysis suggests that a higher dietary inflammatory potential is independently associated with an increased risk of cognitive impairment. These findings underscore the potential impact of dietary inflammation on cognitive health and highlight the need for dietary strategies to mitigate cognitive decline risk.
Collapse
Affiliation(s)
- Boyu Fang
- The Second Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zhipeng Wang
- The Second Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Guangxian Nan
- The Second Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
24
|
van Setten A, Uleman JF, Melis RJF, Lawlor B, Riksen NP, Claassen JAHR, de Heus RAA. No association between markers of systemic inflammation and endothelial dysfunction with Alzheimer's disease progression: a longitudinal study. GeroScience 2025; 47:1093-1104. [PMID: 39085534 PMCID: PMC11872860 DOI: 10.1007/s11357-024-01294-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
INTRODUCTION Systemic inflammation and endothelial dysfunction are potentially modifiable factors implicated in Alzheimer's disease (AD), which offer potential therapeutic targets to slow disease progression. METHODS We investigated the relationship between baseline circulating levels of inflammatory (TNF-α, IL-1ß) and endothelial cell markers (VCAM-1, ICAM-1, E-selectin) and 18-month cognitive decline (ADAS-cog12) in 266 mild-to-moderate AD patients from the NILVAD study. We employed individual growth models to examine associations, potential mediation, and interaction effects while adjusting for confounders. RESULTS The average increase in ADAS-cog12 scores over all patients was 8.1 points in 18 months. No significant association was found between the markers and the rate of cognitive decline. Mediation analysis revealed no mediating role for endothelial cell markers, and interaction effects were not observed. DISCUSSION Our results do not support the role of systemic inflammation or endothelial dysfunction in progression in persons with AD.
Collapse
Affiliation(s)
- Arne van Setten
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen F Uleman
- Copenhagen Health Complexity Center, Department of Public Health, University of Copenhagen, Oster Farimagsgade 5, 1353, Copenhagen K, Denmark.
| | - René J F Melis
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Brian Lawlor
- Global Brain Health Institute, Trinity College, Dublin, Ireland
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Rianne A A de Heus
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Primary and Community Care, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
25
|
Shateri S, Khatami SH, Haghbin Toutounchi A, Rajaei S, Mahdavi M, Mahmoodi Baram S, Shahidi GA, Habibi AH, Aghamollaii V, Ghlichnia B, Safakish L, Doagoo A, Salmani F, Tafakhori A, Keramatinia A, Shahmohammadi MR, Karima S. Plasma cytokines profile in patients with Alzheimer's and Parkinson's Disease: a comparative study in terms of inflammation. Int J Neurosci 2025; 135:158-167. [PMID: 38064237 DOI: 10.1080/00207454.2023.2292951] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/05/2023] [Accepted: 12/02/2023] [Indexed: 02/04/2025]
Abstract
BACKGROUND Neurodegenerative disorders such as Alzheimer's and Parkinson's disease inflict economic and health burdens on societies. Alzheimer's disease (AD), the most prevalent form of dementia, is accompanied by progressive degradation of memory, decision-making, and judgment. Parkinson's disease (PD) is characterized by resting tremor, rigidity, bradykinesia, and loss of balance. Extensive research has pinpointed inflammation as a cause of the onset and progression of both diseases. However, it has not been confirmed which one is more formidable in terms of inflammation. METHODS To assess the extent of inflammation that is implicated in AD and PD and answer the question of which one is more inflammatory, serum levels of inflammatory biomarkers, including cytokines, chemokines, and prostaglandin E2 (PEG2), were measured in AD and PD patients as well as a healthy group. RESULTS Our results showed a significant increase in IL-1α, IL-1β, IL-4, IL-6, IL-10, IL-12p70, IP-10, MCP-1, PEG2, and TNF-α in AD and PD patients compared with the control. Interestingly, IFN-γ did not manifest any significant difference in AD or PD patients compared with the control. CONCLUSION As a hallmark of our results, it could be inferred that inflammation, as the underlying etiological cause, plays a more crucial role in PD compared with AD. Based on our results, it is proposed that anti-inflammatory remedies would be putatively more effective in PD rather than AD.
Collapse
Affiliation(s)
- Somayeh Shateri
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Alireza Haghbin Toutounchi
- Department of General Surgery, Imam Hosein Medical and Educational Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shima Rajaei
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
- Clinical Study Department, Behbalin Inc, Tehran, Iran
| | - Meisam Mahdavi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Somayeh Mahmoodi Baram
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
- Clinical Study Department, Behbalin Inc, Tehran, Iran
| | - Gholam-Ali Shahidi
- Neurology Department, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Amir Hossein Habibi
- Neurology Department, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Vajiheh Aghamollaii
- Neurology Department, Roozbeh Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Babak Ghlichnia
- Department of Neurology, School of Medicine, Iranian Center of Neurological Research, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Lily Safakish
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Alireza Doagoo
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Farzaneh Salmani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Abbas Tafakhori
- Department of Neurology, School of Medicine, Iranian Center of Neurological Research, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Aliasghar Keramatinia
- Department of Community Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Shahmohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| |
Collapse
|
26
|
Komal K, Ghosh R, Sil D, Sharma R, Kumar S, Pandey P, Kumar M. Advancements in nose-to-brain drug targeting for Alzheimer's disease: a review of nanocarriers and clinical insights. Inflammopharmacology 2025; 33:605-626. [PMID: 39776027 DOI: 10.1007/s10787-024-01636-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
Alzheimer's disease (AD) is a type of neurodegenerative disease that describes cognitive decline and memory loss resulting in disability in movement, memory, speech etc. Which first affects the hippocampal and entorhinal cortex regions of brain. Pathogenesis of AD depends on Amyloid-β, hyper-phosphorylation of tau protein, mitochondrial dysfunction, cholinergic hypothesis and oxidative stress. In comparison with males, females are more prone to AD due to reduced estrogen level. Some of the FDA-approved drugs and their conventional formulations available in the market are discussed in this review. Nose-to-brain delivery system provides the target specific drug delivery via olfactory and trigeminal nerve (active and passive drug targeting strategies) and bypassing the Blood Brain Barrier. Mucoadhesive agents and permeation enhancers are mostly utilized to enhance the retention time and bioavailability of the drugs. Liposomes, niosomes, cubosomes, solid lipid nanoparticles, nanoemulsions, micelles, and many more nanocarriers for nose-to-brain delivery of drugs are also described thoroughly in this review. It also covers the clinical trials and patents for nose-to-brain delivery. In this article, we investigate the nose-to-brain pathways for AD treatment strategies.
Collapse
Affiliation(s)
- Kumari Komal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Debayan Sil
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rohit Sharma
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Prachi Pandey
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
27
|
Crine V, Papenberg G, Johansson J, Boraxbekk CJ, Wåhlin A, Lindenberger U, Lövdén M, Riklund K, Bäckman L, Nyberg L, Karalija N. Associations between inflammation and striatal dopamine D2-receptor availability in aging. J Neuroinflammation 2025; 22:24. [PMID: 39885603 PMCID: PMC11783874 DOI: 10.1186/s12974-025-03355-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Normal brain aging is associated with dopamine decline, which has been linked to age-related cognitive decline. Factors underlying individual differences in dopamine integrity at older ages remain, however, unclear. Here we aimed at investigating: (i) whether inflammation is associated with levels and 5-year changes of in vivo dopamine D2-receptor (DRD2) availability, (ii) if DRD2-inflammation associations differ between men and women, and (iii) whether inflammation and cerebral small-vessel disease (white-matter lesions) serve as two independent predictors of DRD2 availability. METHODS Analyses were performed in a sample of healthy adults > 60 years assessed at two measurement occasions separated by 5 years. At both occasions, DRD2 availability was estimated by 11C-raclopride PET, and white-matter lesions by MRI. Inflammation was assessed by two C-reactive protein-associated DNA methylation scores at study baseline. RESULTS Individuals with higher DNA methylation scores at baseline showed reduced striatal DRD2 availability. An interaction was found between DNA methylation scores and sex in relation to striatal DRD2 availability, such that associations were found in men but not in women. DNA methylation scores at study entrance were not significantly associated with 5-year striatal DRD2 decline rates. No significant association was found between DNA methylation scores and white-matter lesions, but higher scores as well as higher lesion burden were independently associated with reduced striatal DRD2 availability in men. CONCLUSIONS These findings suggest negative associations between one proxy of inflammation and DRD2 availability in older adults, selectively for men who had higher DNA methylation scores. Future studies should investigate other inflammatory markers in relation to dopamine integrity.
Collapse
Affiliation(s)
- Vanessa Crine
- Department of Medical and Translational Biology, Umeå university, Umeå, 901 87, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Goran Papenberg
- Aging Research Center, Karolinska Institute and Stockholm University, Stockholm, Sweden
| | - Jarkko Johansson
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Carl-Johan Boraxbekk
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
- Institute for Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Sports Medicine Copenhagen (ISMC), Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Anders Wåhlin
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
- Department of Applied Physics and Electronics, Umeå University, Umeå, Sweden
| | - Ulman Lindenberger
- Center for Lifeorgdivision Psychology, Max Planck Institute for Human Development, Berlin, Germany
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany and London, UK
| | - Martin Lövdén
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden
| | - Katrine Riklund
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Lars Bäckman
- Aging Research Center, Karolinska Institute and Stockholm University, Stockholm, Sweden
| | - Lars Nyberg
- Department of Medical and Translational Biology, Umeå university, Umeå, 901 87, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Nina Karalija
- Department of Medical and Translational Biology, Umeå university, Umeå, 901 87, Sweden.
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden.
| |
Collapse
|
28
|
Kinderlehrer DA. Mushrooms, Microdosing, and Mental Illness: The Effect of Psilocybin on Neurotransmitters, Neuroinflammation, and Neuroplasticity. Neuropsychiatr Dis Treat 2025; 21:141-155. [PMID: 39897712 PMCID: PMC11787777 DOI: 10.2147/ndt.s500337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025] Open
Abstract
The incidence of mental health disorders is increasing worldwide. While there are multiple factors contributing to this problem, neuroinflammation underlies a significant subset of psychiatric conditions, particularly major depressive and anxiety disorders. Anti-inflammatory interventions have demonstrated benefit in these conditions. Psilocin, the active ingredient of mushrooms in the Psilocybe genus, is both a potent serotonin agonist and anti-inflammatory agent, increases neuroplasticity, and decreases overactivity in the default mode network. Studies using hallucinogenic doses of psilocin under the supervision of a therapist/guide have consistently demonstrated benefits to individuals with depression and end-of-life anxiety. Microdosing psilocybin in sub-hallucinogenic doses has also demonstrated benefit in mood disorders, and may offer a safe, less expensive, and more available alternative to full doses of psilocybin for mood disorders, as well as for other medical conditions in which inflammation is the principal pathophysiology.
Collapse
|
29
|
Thomas A, Guo J, Reyes-Dumeyer D, Sanchez D, Scarmeas N, Manly JJ, Brickman AM, Lantigua RA, Mayeux R, Gu Y. Inflammatory biomarkers profiles and cognition among older adults. Sci Rep 2025; 15:2265. [PMID: 39824904 PMCID: PMC11748720 DOI: 10.1038/s41598-025-86309-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025] Open
Abstract
Inflammation plays a major role in cognitive aging. Most studies on peripheral inflammation and cognitive aging focused on selected major inflammatory biomarkers. However, inflammatory markers are regulated and influenced by each other, and it is therefore important to consider a more comprehensive panel of markers to better capture diverse immune pathways and characterize the overall inflammatory profile of individuals. We explored 23 circulating inflammatory biomarkers using data from 1,743 participants without dementia (≥ 65 years-old) from the community-based, multiethnic Washington Heights Inwood Columbia Aging Project. Using principal component analysis (PCA), we developed six inflammatory profiles (PC-1 to PC-6) based on these 23 biomarkers and tested the association of resulting inflammatory profile with cognitive decline, over up to 12 years of follow-up. PC-1 described a pro-inflammatory profile characterized by high positive loadings for pro-inflammatory biomarkers. A higher PC-1 score was associated with lower baseline cognitive performances. No association of this profile with cognitive decline was observed in longitudinal analysis. However, PC-5 characterized by high PDGF-AA and RANTES was associated with a faster cognitive decline. Among older adults, a circulating pro-inflammatory immune profile is associated with lower baseline cognitive performance, and some specific pro-inflammatory cytokines might be associated with faster cognitive decline.
Collapse
Affiliation(s)
- Aline Thomas
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Jing Guo
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Danurys Sanchez
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Nikolaos Scarmeas
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY, 10032, USA
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Jennifer J Manly
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY, 10032, USA
| | - Adam M Brickman
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY, 10032, USA
| | - Rafael A Lantigua
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY, 10032, USA
| | - Richard Mayeux
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY, 10032, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Yian Gu
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY, 10032, USA.
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA.
- Departments of Neurology and Epidemiology, Taub Institute, and Sergievsky Center, Columbia University Irving Medical Center, 622 W 168Th St., New York, NY, 10032, USA.
| |
Collapse
|
30
|
Guo Y, Zhao J, Liu X, Lu P, Liang F, Wang X, Wu J, Hai Y. Ghrelin Induces Ferroptosis Resistance and M2 Polarization of Microglia to Alleviate Neuroinflammation and Cognitive Impairment in Alzheimer's Disease. J Neuroimmune Pharmacol 2025; 20:6. [PMID: 39797928 DOI: 10.1007/s11481-024-10165-3] [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: 07/26/2024] [Accepted: 11/29/2024] [Indexed: 01/13/2025]
Abstract
Microglial polarization and ferroptosis are important pathological features in Alzheimer's disease (AD). Ghrelin, a brain-gut hormone, has potential neuroprotective effects in AD. This study aimed to explore the potential mechanisms by which ghrelin regulates the progression of AD, as well as the crosstalk between microglial polarization and ferroptosis. Mouse BV2 microglial cells and male mice were treated with beta-amyloid (Aβ) (1-42) to simulate the AD environment. Microglia ferroptosis was measured by detecting levels of ferroptosis-related proteins (SLC7A11, GPX4, FTL1, and FTH1), metabolic markers (ROS, MDA, GSH, SOD), and observing mitochondrial morphological changes. Microglial polarization was evaluated by measuring levels of inflammatory markers and surface markers. The impact of ghrelin on Aβ1-42-exposed microglia was assessed by coupling with the ferroptosis activator Erastin. Cognitive impairment in AD mice was evaluated through behavioral tests. Tissue staining was applied to determine neuronal damage. In Aβ1-42-exposed microglia, ghrelin upregulated the protein expression of SLC7A11, GPX4, FTL1 and FTH1, reduced ROS and MDA levels, and elevated GSH and SOD levels through the BMP6/SMAD1 pathway. Ghrelin alleviated mitochondrial structural damage. Additionally, ghrelin reduced levels of pro-inflammatory factors and CD86, while increasing levels of anti-inflammatory factors and CD206. Erastin reversed the effects of ghrelin on ferroptosis and phenotypic polarization in Aβ1-42-exposed microglia. In AD mice, ghrelin ameliorated abnormal behavior, neuroinflammation, and plaque deposition. Ghrelin attenuated iNOS/IBA1-positive expression and enhanced Arg-1/IBA1-positive expression in the hippocampus. Ghrelin induces microglial M2 polarization by inhibiting microglia ferroptosis, thereby alleviating neuroinflammation. Our results indicate that ghrelin may serve as a promising potential agent for treating cognitive impairment in AD.
Collapse
Affiliation(s)
- Yaoxue Guo
- Department of Clinical Pharmacy, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Junli Zhao
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Xing Liu
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Pu Lu
- Oncology Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Furu Liang
- Department of Neurology, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Xueyan Wang
- Oncology Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Jing Wu
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China
| | - Yan Hai
- Pharmacy Department, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, China.
- Baotou Central Hospital, 61 Huancheng Road, Donghe District, Baotou, 014040, Inner Mongolia, China.
| |
Collapse
|
31
|
Volloch V, Rits-Volloch S. Production of Amyloid-β in the Aβ-Protein-Precursor Proteolytic Pathway Is Discontinued or Severely Suppressed in Alzheimer's Disease-Affected Neurons: Contesting the 'Obvious'. Genes (Basel) 2025; 16:46. [PMID: 39858593 PMCID: PMC11764795 DOI: 10.3390/genes16010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
A notion of the continuous production of amyloid-β (Aβ) via the proteolysis of Aβ-protein-precursor (AβPP) in Alzheimer's disease (AD)-affected neurons constitutes both a cornerstone and an article of faith in the Alzheimer's research field. The present Perspective challenges this assumption. It analyses the relevant empirical data and reaches an unexpected conclusion, namely that in AD-afflicted neurons, the production of AβPP-derived Aβ is either discontinued or severely suppressed, a concept that, if proven, would fundamentally change our understanding of the disease. This suppression, effectively self-suppression, occurs in the context of the global inhibition of the cellular cap-dependent protein synthesis as a consequence of the neuronal integrated stress response (ISR) elicited by AβPP-derived intraneuronal Aβ (iAβ; hence self-suppression) upon reaching certain levels. Concurrently with the suppression of the AβPP proteolytic pathway, the neuronal ISR activates in human neurons, but not in mouse neurons, the powerful AD-driving pathway generating the C99 fragment of AβPP independently of AβPP. The present study describes molecular mechanisms potentially involved in these phenomena, propounds novel approaches to generate transgenic animal models of AD, advocates for the utilization of human neuronal cells-based models of the disease, makes verifiable predictions, suggests experiments designed to validate the proposed concept, and considers its potential research and therapeutic implications. Remarkably, it opens up the possibility that the conventional production of AβPP, BACE enzymes, and γ-secretase components is also suppressed under the neuronal ISR conditions in AD-affected neurons, resulting in the dyshomeostasis of AβPP. It follows that whereas conventional AD is triggered by AβPP-derived iAβ accumulated to the ISR-eliciting levels, the disease, in its both conventional and unconventional (triggered by the neuronal ISR-eliciting stressors distinct from iAβ) forms, is driven not (or not only) by iAβ produced in the AβPP-independent pathway, as we proposed previously, but mainly, possibly exclusively, by the C99 fragment generated independently of AβPP and not cleaved at the γ-site due to the neuronal ISR-caused deficiency of γ-secretase (apparently, the AD-driving "substance X" predicted in our previous study), a paradigm consistent with a dictum by George Perry that Aβ is "central but not causative" in AD. The proposed therapeutic strategies would not only deplete the driver of the disease and abrogate the AβPP-independent production of C99 but also reverse the neuronal ISR and ameliorate the AβPP dyshomeostasis, a potentially significant contributor to AD pathology.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
32
|
Counil H, Silva RO, Rabanel J, Zaouter C, Haddad M, Ben Khedher MR, Brambilla D, Fülöp T, Patten SA, Ramassamy C. Brain penetration of peripheral extracellular vesicles from Alzheimer's patients and induction of microglia activation. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70027. [PMID: 39830834 PMCID: PMC11740088 DOI: 10.1002/jex2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/13/2024] [Accepted: 12/08/2024] [Indexed: 01/22/2025]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative pathology. Brain-derived extracellular vesicles (EVs) have been demonstrated to be implicated in AD pathogenesis by facilitating the propagation of Tau, amyloid-β and inflammatory cytokines. However, the impact of peripheral EVs (pEVs) in AD pathogenesis remains poorly investigated. The objective of our study was to compare the passage of pEVs from adults, cognitively healthy elderly, and AD patients through the blood-brain barrier (BBB), to evaluate their uptake in the brain and to assess their impact on the microglia activity using in vitro and in vivo models. To this end, pEVs were enriched, characterized, and fluorescently labelled. The passage of pEVs through the endothelial bEnd.3 cells was studied in a Transwell device with either neuronal or microglia cells seeded at the bottom of the well. Following the internalization of pEVs from AD patients, microglia adopted an amoeboid morphology and released a heightened level of pro-inflammatory cytokine IL-6. To further assess their in vivo transport across the BBB, pEVs were injected into the blood circulation of 2-days post-fertilization Tg(flk1:EGFP) zebrafish. The biodistribution of pEVs was monitored at 1 and 24 h post-injection using confocal microscopy. We demonstrated that pEVs traverse the BBB by transcytosis and subsequently diffuse progressively into the brain. pEVs were then internalized by neuronal and radial glial cells as seen in Tg(huc:EGFP) and Tg(gfap:EGFP) zebrafish, respectively. Additional experiments were performed with the intrahippocampal injection of pEVs in the mouse, indicating their spreading throughout the brain and their uptake by neuronal and glial cells. These findings contribute to novel insights into the fate of pEVs following their passage through the BBB in vitro and in vivo, and demonstrate for the first time that pEVs from AD patients affect microglia activity. This suggests a potential mechanism through which peripheral tissue cues may contribute to AD pathogenesis.
Collapse
Affiliation(s)
- Hermine Counil
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
| | | | - Jean‐Michel Rabanel
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
- School of Pharmaceutical Sciences, Faculty of MedecineUniversity of OttawaOttawaOntarioCanada
| | | | - Mohamed Haddad
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
| | - Mohamed Raâfet Ben Khedher
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
- Higher Institute of Biotechnology of BejaUniversity of JendoubaBejaTunisia
| | - Davide Brambilla
- Université de MontréalFaculté de Pharmacie, Pavillon Jean‐CoutuMontréalQuebecCanada
| | - Tamas Fülöp
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health SciencesUniversity of SherbrookeSherbrookeQuebecCanada
| | | | | |
Collapse
|
33
|
Seady M, Schirmbeck G, Taday J, Fróes FT, Baú JV, Jantsch J, Guedes RP, Gonçalves CA, Leite MC. Curcumin attenuates neuroinflammatory damage induced by LPS: Implications for the role of S100B. J Nutr Biochem 2025; 135:109768. [PMID: 39278425 DOI: 10.1016/j.jnutbio.2024.109768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/24/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Inflammation is a common feature of neurological disorders that alters cell function in microglia and astrocytes as well as other neuronal cell types. Astrocytes modulate blood flow, regulate glutamate metabolism, and exert antioxidant protection. When responding to inflammatory damage, astrocytes enhance immune cell infiltration and amplify inflammatory responses via the upregulation of cytokine production. Several molecules have been proposed to attenuate neuroinflammation and control neurological diseases. Curcumin gained attention due to its capacity to cross the blood-brain barrier and its well-described anti-inflammatory and antioxidant activities. Our study aimed to understand if oral curcumin administration could protect against central nervous system inflammatory damage induced by intracerebroventricular injection of LPS while focusing on astrocyte function. Despite its poor bioavailability, we found that curcumin reaches the central nervous system, prevents the locomotory damage caused by LPS, and reduces inflammatory signaling via IL-1β and COX-2. Furthermore, we observed that curcumin was protective against LPS-induced S100B secretion in the cerebrospinal fluid and GSH reduction in the hippocampal tissue. However, curcumin could not protect the animals from anhedonia, assessed by the sucrose preference test, and weight loss induced by LPS. Our results indicate that oral curcumin administration exerts a protective anti-inflammatory action in the central nervous system, attenuating the sickness behavior induced by ICV LPS. This work demonstrates that curcumin has an important modulative effect on astrocytes, thus suggesting that astrocytes are critical to the anti-inflammatory effects of curcumin.
Collapse
Affiliation(s)
- Marina Seady
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gabriel Schirmbeck
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéssica Taday
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Telles Fróes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéfeli Vasques Baú
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jeferson Jantsch
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marina Concli Leite
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
34
|
Valiukas Z, Tangalakis K, Apostolopoulos V, Feehan J. Microglial activation states and their implications for Alzheimer's Disease. J Prev Alzheimers Dis 2025; 12:100013. [PMID: 39800461 DOI: 10.1016/j.tjpad.2024.100013] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Alzheimer's Disease (AD) is a chronic neurodegenerative disorder characterized by the accumulation of toxic amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFTs) of tau protein in the brain. Microglia, key immune cells of the central nervous system, play an important role in AD development and progression, primarily through their responses to Aβ and NFTs. Initially, microglia can clear Aβ, but in AD, chronic activation overwhelms protective mechanisms, leading to sustained neuroinflammation that enhances plaque toxicity, setting off a damaging cycle that affects neurons, astrocytes, cerebral vasculature, and other microglia. Current AD treatments have been largely ineffective, though emerging immunotherapies focusing on plaque removal show promise, but often overlook the role of neuroinflammation. Activated microglia display a complex range of phenotypes that can be broadly broken into pro- or anti-inflammatory states, although this dichotomy does not describe the significant overlap between states. Aβ can strongly induce inflammatory activity, triggering the production of reactive oxygen species, inflammatory cytokines (e.g., TNF-α, IL-1β, IL-6), synapse engulfment, blood-brain barrier compromise, and impaired Aβ clearance. These processes contribute to neural tissue loss, manifesting as cognitive decline such as impaired executive function and memory. Conversely, anti-inflammatory activation exerts neuroprotective effects by suppressing inflammatory pathways and releasing neurotrophic factors that aid neuron repair and protection. Induction of anti-inflammatory states may offer a dual therapeutic approach to address both neuroinflammation and plaque accumulation in AD. This approach suggests potential strategies to modulate microglial phenotypes, aiming to restore neuroprotective functions and mitigate disease progression by simultaneously targeting inflammation and plaque pathology.
Collapse
Affiliation(s)
- Zachary Valiukas
- Institute for Health and Sport, Victoria University, 70/104 Ballarat Road, Footscray VIC 3011, Australia
| | - Kathy Tangalakis
- First Year College, Victoria University, 70/104 Ballarat Road, Footscray VIC 3011, Australia
| | - Vasso Apostolopoulos
- School of Health and Biomedical Sciences, RMIT University, 220 3-5 Plenty Road, Bundoora VIC 3082, Australia.
| | - Jack Feehan
- School of Health and Biomedical Sciences, RMIT University, 220 3-5 Plenty Road, Bundoora VIC 3082, Australia.
| |
Collapse
|
35
|
Ko YS, Ryu YK, Han S, Park HJ, Choi M, Kim BC, Jeong HS, Jang S, Jo J, Lee S, Choi WS, Cho HH. Hearing modulation affects Alzheimer's disease progression linked to brain inflammation: a study in mouse models. Mol Med 2024; 30:276. [PMID: 39725872 PMCID: PMC11670416 DOI: 10.1186/s10020-024-01040-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Recent studies have identified hearing loss (HL) as a primary risk factor for Alzheimer's disease (AD) onset. However, the mechanisms linking HL to AD are not fully understood. This study explored the effects of drug-induced hearing loss (DIHL) on the expression of proteins associated with AD progression in mouse models. METHODS DIHL was induced in 5xFAD and Tg2576 mice aged 3 to 3.5 weeks using kanamycin (700 mg/kg, subcutaneous) and furosemide (600 mg/kg, intraperitoneal). The accumulation and expression of beta-amyloid (Aβ), ionized calcium-binding adaptor molecule 1 (Iba1), and glial fibrillary acidic protein (GFAP) were measured through immunohistochemistry and immunoblotting. Additionally, the expression of proteins involved in the mammalian target of rapamycin (mTOR) pathway, including downstream effectors p70 ribosomal S6 kinase (p70S6K) and S6, as well as proinflammatory cytokines, was analyzed. RESULTS Compared to control conditions, HL led to a significant increase in the accumulation of Aβ in the hippocampus and cortex. Elevated levels of neuroinflammatory markers, including Iba1 and GFAP, as well as proinflammatory cytokines such as interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α), were observed. Moreover, DIHL enhanced phosphorylation of mTOR, p70S6K, and S6, indicating activation of the mTOR pathway. CONCLUSIONS HL significantly increases Aβ accumulation in the brain. Furthermore, HL activates astrocytes and microglia, leading to increased neuroinflammation and thereby accelerating AD progression. These findings strongly suggest that HL contributes autonomously to neuroinflammation, highlighting the potential for early intervention in HL to reduce AD risk.
Collapse
Affiliation(s)
- Yoo-Seung Ko
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jaebong-Ro, Dong-Gu, Gwangju, 61469, Republic of Korea
| | - Young-Kyoung Ryu
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jaebong-Ro, Dong-Gu, Gwangju, 61469, Republic of Korea
| | - Sujin Han
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jaebong-Ro, Dong-Gu, Gwangju, 61469, Republic of Korea
| | - Hyung Joon Park
- Department of Biochemistry, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Munyoung Choi
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jaebong-Ro, Dong-Gu, Gwangju, 61469, Republic of Korea
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Medical School & Hospital, Gwangju, 61469, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun-Gun, Jeollanamdo, 58128, Republic of Korea
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun-Gun, Jeollanamdo, 58128, Republic of Korea
| | - Jihoon Jo
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Sungsu Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jaebong-Ro, Dong-Gu, Gwangju, 61469, Republic of Korea
| | - Won-Seok Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea.
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jaebong-Ro, Dong-Gu, Gwangju, 61469, Republic of Korea.
| |
Collapse
|
36
|
Ciaglia T, Miranda MR, Di Micco S, Vietri M, Smaldone G, Musella S, Di Sarno V, Auriemma G, Sardo C, Moltedo O, Pepe G, Bifulco G, Ostacolo C, Campiglia P, Manfra M, Vestuto V, Bertamino A. Neuroprotective Potential of Indole-Based Compounds: A Biochemical Study on Antioxidant Properties and Amyloid Disaggregation in Neuroblastoma Cells. Antioxidants (Basel) 2024; 13:1585. [PMID: 39765912 PMCID: PMC11673510 DOI: 10.3390/antiox13121585] [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: 11/12/2024] [Revised: 12/15/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
Based on the established neuroprotective properties of indole-based compounds and their significant potential as multi-targeted therapeutic agents, a series of synthetic indole-phenolic compounds was evaluated as multifunctional neuroprotectors. Each compound demonstrated metal-chelating properties, particularly in sequestering copper ions, with quantitative analysis revealing approximately 40% chelating activity across all the compounds. In cellular models, these hybrid compounds exhibited strong antioxidant and cytoprotective effects, countering reactive oxygen species (ROS) generated by the Aβ(25-35) peptide and its oxidative byproduct, hydrogen peroxide, as demonstrated by quantitative analysis showing on average a 25% increase in cell viability and a reduction in ROS levels to basal states. Further analysis using thioflavin T fluorescence assays, circular dichroism, and computational studies indicated that the synthesized derivatives effectively promoted the self-disaggregation of the Aβ(25-35) fragment. Taken together, these findings suggest a unique profile of neuroprotective actions for indole-phenolic derivatives, combining chelating, antioxidant, and anti-aggregation properties, which position them as promising compounds for the development of multifunctional agents in Alzheimer's disease therapy. The methods used provide reliable in vitro data, although further in vivo validation and assessment of blood-brain barrier penetration are needed to confirm therapeutic efficacy and safety.
Collapse
Affiliation(s)
- Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Simone Di Micco
- European Biomedical Research Institute of Salerno (EBRIS), Via Salvatore de Renzi 50, 84125 Salerno, Italy;
| | - Mariapia Vietri
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Gerardina Smaldone
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Giulia Auriemma
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Carla Sardo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Michele Manfra
- Department of Health Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (T.C.); (M.R.M.); (M.V.); (G.S.); (S.M.); (V.D.S.); (G.A.); (C.S.); (O.M.); (G.P.); (G.B.); (C.O.); (P.C.); (A.B.)
| |
Collapse
|
37
|
Ali A, Chaudhary A, Sharma A, Siddiqui N, Anurag, Parihar VK. Exploring role of citrus fruits in comorbid neurodegenerative disorders associated with psoriasis. Metab Brain Dis 2024; 40:62. [PMID: 39671136 DOI: 10.1007/s11011-024-01479-5] [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: 05/06/2024] [Accepted: 11/09/2024] [Indexed: 12/14/2024]
Abstract
A neurodegenerative illness is a disorder in which the brain and/or spinal cord's neurons, or nerve cells, gradually deteriorate and disappear. These illnesses often get worse with time and can seriously affect movement, cognition, and other neurological functions. Psoriasis is a long-term autoimmune skin condition marked by fast skin cell growth that results in red, elevated areas coated in silvery-white scales. It can affect several body parts, such as the elbows, knees, scalp, and lower back, and it is not communicable. The build-up of amyloid beta [Aβ] protein is linked to elevated levels of reactive oxygen species (ROS) (Kim et al. 2020). These ROS can trigger multiple pathways, including MAPK, NFkB, JAK/STAT, and interleukin 1 beta (IL-1β), ultimately playing a role in the development of neurodegenerative illnesses like Alzheimer's disease (AD) and psoriasis. People who have psoriasis are more likely to acquire AD, as psoriasis is a chronic inflammatory skin condition that is genetically connected. Because of the antioxidants and anti-inflammatory properties of citrus fruits neurodegenerative and psoriasis disease may be prevented. The neuroprotective action of bioactives in citrus fruits involves the inhibition of inflammation through the control of p38 mitogen-activated protein kinase (MAPK) and the activation of nuclear factor erythroid 2-related factor 2 (Nrf2). Due to their immunomodulatory and anti-inflammatory qualities, polyphenols may be able to control the immune response in psoriasis. We performed a thorough review in order to investigate for the first time to understand the role of citrus fruits in comorbid neurodegenerative disorders associated with psoriasis. For better understanding into the possible applications of citrus fruits in treating psoriasis and neurodegenerative disease would require additional studies focusing directly on the relationship between citrus fruits consumption in managing neurodegenerative and psoriasis disease.
Collapse
Affiliation(s)
- Adil Ali
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology (MIET), Meerut, 250005, India
| | - Ankit Chaudhary
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology (MIET), Meerut, 250005, India
| | - Alok Sharma
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology (MIET), Meerut, 250005, India.
| | - Nazia Siddiqui
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology (MIET), Meerut, 250005, India
| | - Anurag
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) Hajipur, Hajipur, 844102, India
| | - Vipan Kumar Parihar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology (MIET), Meerut, 250005, India
| |
Collapse
|
38
|
Bacherini D, Maggi L, Faraldi F, Sodi A, Vannozzi L, Mazzoni A, Capone M, Virgili G, Vicini G, Falsini B, Cosmi L, Viggiano P, Rizzo S, Annunziato F, Giansanti F, Liotta F. CD3+CD4-CD8- Double-Negative Lymphocytes Are Increased in the Aqueous Humor of Patients with Retinitis Pigmentosa: Their Possible Role in Mediating Inflammation. Int J Mol Sci 2024; 25:13163. [PMID: 39684872 DOI: 10.3390/ijms252313163] [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: 10/20/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Recently, evidence has supported a significant role for immune and oxidative-mediated damage underlying the pathogenesis of different types of retinal diseases, including retinitis pigmentosa (RP). Our study aimed to evaluate the presence of immune cells and mediators in patients with RP using flow cytometric analysis of peripheral blood (PB) and aqueous humor (AH) samples. We recruited 12 patients with RP and nine controls undergoing cataract surgery. Flow cytometric analysis of PB and AH samples provided a membrane staining that targeted surface molecules (CD14, CD16, CD19, CD3, CD4, CD8, and CD161) identifying monocytes, natural killer (NK) cells, B cells, T cells, and T subpopulations, respectively. Moreover, lymphocytes were polyclonally stimulated to evaluate cytokine (CK) production at single-cell level. The circulating immune cell distribution was comparable between patients with RP and controls. Conversely, in the AH of controls we could detect no cells, while in the RP AH samples we found infiltrating leukocytes, consisting of T (CD3+), B (CD19+), NK (CD16+CD3-) cells, and monocytes (CD14+). In patients with RP, the frequency of most infiltrating immune cell populations was similar between the AH and PB. However, among T cell subpopulations, the frequency of CD3+CD4+ T cells was significantly lower in the RP AH compared to RP PB, whereas CD3+CD4-CD8- double-negative (DN) T cells were significantly higher in the RP AH compared to RP PB. Cytokine production analysis revealed a trend toward an increased frequency of CD3+CD8-CD161+IFN-ɣ-producing cells and a decreased frequency of CD3+CD8+IL-4-producing cells in the RP AH compared to RP PB. The detection of immune cells, particularly DN T cells, and a Th1-skewed phenotype in RP AH suggests immune-mediated and inflammatory mechanisms in the disease.
Collapse
Affiliation(s)
- Daniela Bacherini
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Francesco Faraldi
- Ophthalmology Unit, Surgical Department, A.O. Ordine Mauriziano, 10128 Turin, Italy
| | - Andrea Sodi
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Lorenzo Vannozzi
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Gianni Virgili
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Giulio Vicini
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Benedetto Falsini
- UOC Oculistica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Head and Neck and Sensory Organs, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Pasquale Viggiano
- Department of Translational Biomedicine Neuroscience, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Stanislao Rizzo
- UOC Oculistica, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Head and Neck and Sensory Organs, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Consiglio Nazionale delle Ricerche, Istituto di Neuroscienze, 56124 Pisa, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Fabrizio Giansanti
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139 Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
39
|
Pasupalak JK, Rajput P, Gupta GL. Gut microbiota and Alzheimer's disease: Exploring natural product intervention and the Gut-Brain axis for therapeutic strategies. Eur J Pharmacol 2024; 984:177022. [PMID: 39362390 DOI: 10.1016/j.ejphar.2024.177022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/14/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Numerous studies conducted over the last ten years have shown a strong correlation between the gut microbiota and the onset and progression of Alzheimer's disease (AD). However, the exact underlying mechanism is still unknown. An ongoing communication mechanism linking the gut and the brain is highlighted by the term "microbiota-gut-brain axis," which was originally coined the "gut-brain axis." Key metabolic, endocrine, neurological, and immunological mechanisms are involved in the microbiota‒gut‒brain axis and are essential for preserving brain homeostasis. Thus, the main emphasis of this review is how the gut microbiota contributes to the development of AD and how various natural products intervene in this disease. The first part of the review provides an outline of various pathways and relationships between the brain and gut microbiota, and the second part provides various mechanisms involved in the gut microbiota and AD. Finally, this review provides knowledge about natural products and their effectiveness in treating gut microbiota-induced AD. AD may be treated in the future by altering the gut microbiota with a customized diet, probiotics/prebiotics, plant products, and natural products. This entails altering the microbiological partners and products (such as amyloid protein) that these partners generate.
Collapse
Affiliation(s)
- Jajati K Pasupalak
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Prabha Rajput
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Girdhari Lal Gupta
- School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India.
| |
Collapse
|
40
|
Gonzalo R, Minguet C, Ortiz AM, Bravo MI, López OL, Boada M, Ruiz A, Costa M. Plasma exchange with albumin replacement for Alzheimer's disease treatment induced changes in serum and cerebrospinal fluid inflammatory mediator levels. Ann Clin Transl Neurol 2024; 11:3280-3291. [PMID: 39476248 DOI: 10.1002/acn3.52235] [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: 05/24/2024] [Revised: 09/09/2024] [Accepted: 10/01/2024] [Indexed: 12/18/2024] Open
Abstract
OBJECTIVE There is extensive literature indicating that inflammatory pathways are affected in Alzheimer's disease (AD). We examined whether plasma exchange with albumin replacement (PE-Alb) can impact the inflammatory status of AD patients and alter the relationship between inflammatory mediators and cognitive measures. METHODS Serum and cerebrospinal fluid (CSF) samples from 142 AD patients participating in the AMBAR trial (14-month schedule of PE-Alb treatment vs. placebo [sham PE-Alb]) were analyzed for changes from baseline for 19 inflammatory mediators (6 inflammatory cytokines, 9 chemokines, and 4 vascular injury indicators) at representative time points across the AMBAR study (lasting effects) as well as in pre- versus post-PE-Alb procedure (acute effects). Association between mediator changes and clinical outcomes reported in the AMBAR study (cognitive, functional, behavioral function, and global change tests) was assessed. RESULTS PE-Alb significantly reduced IFN-γ, eotaxin, MIP-1α and ICAM-1 levels in serum, and eotaxin-3 and MIP-1β levels in CSF, at various time points during treatment (p < 0.05; false discovery rate-corrected). Vascular injury indicators were the mediators mostly affected by post- versus pre-PE-Alb level reduction. Increased serum MIP-1α levels were associated with worsening in ADAS-Cog, CDR-sb, and ADCS-CGIC scores in the placebo group, but not in the PE-Alb-treated group. INTERPRETATION Peripheral intervention could affect AD by reducing inflammatory mediators in both peripheral and central compartments. Changes in MIP-1α due to PE-Alb were associated with changes in clinical outcomes.
Collapse
Affiliation(s)
- Ricardo Gonzalo
- Grifols Scientific Innovation Office, Avinguda de la Generalitat 152-158, Sant Cugat del Vallès, 08174, Barcelona, Spain
| | - Carla Minguet
- Grifols Scientific Innovation Office, Avinguda de la Generalitat 152-158, Sant Cugat del Vallès, 08174, Barcelona, Spain
| | - Ana María Ortiz
- Grifols Scientific Innovation Office, Avinguda de la Generalitat 152-158, Sant Cugat del Vallès, 08174, Barcelona, Spain
| | - María Isabel Bravo
- Grifols Scientific Innovation Office, Avinguda de la Generalitat 152-158, Sant Cugat del Vallès, 08174, Barcelona, Spain
| | - Oscar L López
- Departments of Neurology and Psychiatry, University of Pittsburgh School of Medicine, 811 Kaufmann Medical Building, 3471 Fifth Avenue, Pittsburgh, 15213, Pennsylvania, USA
| | - Mercè Boada
- ACE Alzheimer Centre Barcelona - Universitat Internacional de Catalunya, Gran Via de Carles III, 85 BIS, Barcelona, 08028, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Calle de Melchor Fernández Almagro 3, Fuencarral-El Pardo, Madrid, 28029, Spain
| | - Agustín Ruiz
- ACE Alzheimer Centre Barcelona - Universitat Internacional de Catalunya, Gran Via de Carles III, 85 BIS, Barcelona, 08028, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Calle de Melchor Fernández Almagro 3, Fuencarral-El Pardo, Madrid, 28029, Spain
| | - Montserrat Costa
- Grifols Scientific Innovation Office, Avinguda de la Generalitat 152-158, Sant Cugat del Vallès, 08174, Barcelona, Spain
| |
Collapse
|
41
|
Tallino S, Etebari R, McDonough I, Leon H, Sepulveda I, Winslow W, Bartholomew SK, Perez SE, Mufson EJ, Velazquez R. Assessing the Benefit of Dietary Choline Supplementation Throughout Adulthood in the Ts65Dn Mouse Model of Down Syndrome. Nutrients 2024; 16:4167. [PMID: 39683562 DOI: 10.3390/nu16234167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Down syndrome (DS) is the most common cause of early-onset Alzheimer's disease (AD). Dietary choline has been proposed as a modifiable factor to improve the cognitive and pathological outcomes of AD and DS, especially as many do not reach adequate daily intake levels of choline. While lower circulating choline levels correlate with worse pathological measures in AD patients, choline status and intake in DS is widely understudied. Perinatal choline supplementation (Ch+) in the Ts65Dn mouse model of DS protects offspring against AD-relevant pathology and improves cognition. Further, dietary Ch+ in adult AD models also ameliorates pathology and improves cognition. However, dietary Ch+ in adult Ts65Dn mice has not yet been explored; thus, this study aimed to supply Ch+ throughout adulthood to determine the effects on cognition and DS co-morbidities. METHODS We fed trisomic Ts65Dn mice and disomic littermate controls either a choline normal (ChN; 1.1 g/kg) or a Ch+ (5 g/kg) diet from 4.5 to 14 months of age. RESULTS We found that Ch+ in adulthood failed to improve genotype-specific deficits in spatial learning. However, in both genotypes of female mice, Ch+ significantly improved cognitive flexibility in a reverse place preference task in the IntelliCage behavioral phenotyping system. Further, Ch+ significantly reduced weight gain and peripheral inflammation in female mice of both genotypes, and significantly improved glucose metabolism in male mice of both genotypes. CONCLUSIONS Our findings suggest that adulthood choline supplementation benefits behavioral and biological factors important for general well-being in DS and related to AD risk.
Collapse
Affiliation(s)
- Savannah Tallino
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Rachel Etebari
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Ian McDonough
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Hector Leon
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Isabella Sepulveda
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Wendy Winslow
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Samantha K Bartholomew
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Sylvia E Perez
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Elliott J Mufson
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Ramon Velazquez
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| |
Collapse
|
42
|
Cómitre-Mariano B, Vellila-Alonso G, Segura-Collar B, Mondéjar-Ruescas L, Sepulveda JM, Gargini R. Sentinels of neuroinflammation: the crucial role of myeloid cells in the pathogenesis of gliomas and neurodegenerative diseases. J Neuroinflammation 2024; 21:304. [PMID: 39578808 PMCID: PMC11583668 DOI: 10.1186/s12974-024-03298-y] [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: 03/04/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024] Open
Abstract
The inflammatory processes that drive pathologies of the central nervous system (CNS) are complex and involve significant contributions from the immune system, particularly myeloid cells. Understanding the shared and distinct pathways of myeloid cell regulation in different CNS diseases may offer critical insights into therapeutic development. This review aims to elucidate the mechanisms underlying myeloid cell dysfunction and neuroinflammation in two groups of neurological pathologies with significant social impact and a limited efficacy of their treatments: the most common primary brain tumors -gliomas-, and the most prevalent neurodegenerative disorders -Alzheimer's and Parkinson's disease. Despite their distinct clinical manifestations, these diseases share key pathological features, including chronic inflammation and immune dysregulation. The role of myeloid cells in neuroinflammation has garnered special interest in recent years in both groups, as evidenced by the growing focus on therapeutic research centred on myeloid cells. By examining the cellular and molecular dynamics that govern these conditions, we hope to identify common and unique therapeutic targets that can inform the development of more effective treatments. Recent advances in single-cell technologies have revolutionized our understanding of myeloid cell heterogeneity, revealing diverse phenotypes and molecular profiles across different disease stages and microenvironments. Here, we present a comprehensive analysis of myeloid cell involvement in gliomas, Alzheimer's and Parkinson's disease, with a focus on phenotypic acquisition, molecular alterations, and therapeutic strategies targeting myeloid cells. This integrated approach not only addresses the limitations of current treatments but also suggests new avenues for therapeutic intervention, aimed at modulating the immune landscape to improve patient outcomes.
Collapse
Affiliation(s)
- Blanca Cómitre-Mariano
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
| | - Gabriel Vellila-Alonso
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
| | - Berta Segura-Collar
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
| | - Lucía Mondéjar-Ruescas
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain
| | - Juan M Sepulveda
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.
| | - Ricardo Gargini
- Instituto de Investigación Biomédicas I+12, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.
- Pathology and Neurooncology Unit, Hospital Universitario 12 de Octubre, Av. de Córdoba, S/N, Madrid, 28041, Spain.
| |
Collapse
|
43
|
Kazemi N, Khorasgani MR, Noorbakhshnia M, Razavi SM, Narimani T, Naghsh N. Protective effects of a lactobacilli mixture against Alzheimer's disease-like pathology triggered by Porphyromonas gingivalis. Sci Rep 2024; 14:27283. [PMID: 39516514 PMCID: PMC11549306 DOI: 10.1038/s41598-024-77853-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is one of the pathogens involved in gingival inflammation, which may trigger neuroinflammatory diseases such as Alzheimer's disease (AD). This study aimed to investigate the protective (preventive and treatment) effects of a lactobacilli mixture combining Lactobacillus reuteri PTCC1655, Lactobacillus brevis CD0817, Lacticaseibacillus rhamnosus PTCC1637, and Lactobacillus plantarum PTCC1058 against P. gingivalis-induced gingival inflammation and AD-like pathology in rats. These probiotic strains exhibited cognitive enhancement effects, but this study proposed to assess their activity in a mixture. To propose a probable mechanism for P. gingivalis cognitive impairments, the TEs balance were analyzed in hippocampus and cortex tissues. Animals were divided into five groups: the control, lactobacilli, P. gingivalis, lactobacilli + P. gingivalis (prevention), and P. gingivalis + lactobacilli group (treatment) groups. The behavioral and histopathological changes were compared among them. Finally, The Trace elements (TEs) levels in the hippocampus and cortex tissues were analyzed. The palatal tissue sections of the P. gingivalis infected rats showed moderate inflammation with dense infiltration of inflammatory cells, a limited area of tissue edema, and vascular congestion. Additionally, passive avoidance learning and spatial memory were impaired. Histopathological tests revealed the presence of Aβ-positive cells in the P. gingivalis group. While the Aβ-positive cells decreased in the treatment group, their formation was inhibited in the preventive group. Administration of a mixture of lactobacilli (orally) effectively mitigated the gingival inflammation, Aβ production, and improved learning and memory functions. Moreover, Zn, Cu, and Mn levels in the hippocampus were dramatically elevated by P. gingivalis infection, whereas lactobacilli mixture mitigated these disruptive effects. The lactobacilli mixture significantly prevented the disruptive effects of P. gingivalis on gingival and brain tissues in rats. Therefore, new formulated combination of lactobacilli may be a good candidate for inhibiting the P. gingivalis infection and its subsequent cognitive effects. The current study aimed to evaluate the effects of a lactobacilli mixture to manage the disruptive effects of P. gingivalis infection on memory.
Collapse
Affiliation(s)
- Niloofar Kazemi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammad Rabbani Khorasgani
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Maryam Noorbakhshnia
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Seyed Mohammad Razavi
- Department of Oral and Maxillofacial Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tahmineh Narimani
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Narges Naghsh
- Department of Periodontology, Torabinejad Dental Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
44
|
Tenchov R, Sasso JM, Zhou QA. Alzheimer's Disease: Exploring the Landscape of Cognitive Decline. ACS Chem Neurosci 2024; 15:3800-3827. [PMID: 39392435 PMCID: PMC11587518 DOI: 10.1021/acschemneuro.4c00339] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. The pathology of AD is marked by the accumulation of amyloid beta plaques and tau protein tangles in the brain, along with neuroinflammation and synaptic dysfunction. Genetic factors, such as mutations in APP, PSEN1, and PSEN2 genes, as well as the APOE ε4 allele, contribute to increased risk of acquiring AD. Currently available treatments provide symptomatic relief but do not halt disease progression. Research efforts are focused on developing disease-modifying therapies that target the underlying pathological mechanisms of AD. Advances in identification and validation of reliable biomarkers for AD hold great promise for enhancing early diagnosis, monitoring disease progression, and assessing treatment response in clinical practice in effort to alleviate the burden of this devastating disease. In this paper, we analyze data from the CAS Content Collection to summarize the research progress in Alzheimer's disease. We examine the publication landscape in effort to provide insights into current knowledge advances and developments. We also review the most discussed and emerging concepts and assess the strategies to combat the disease. We explore the genetic risk factors, pharmacological targets, and comorbid diseases. Finally, we inspect clinical applications of products against AD with their development pipelines and efforts for drug repurposing. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding AD, to outline challenges, and to evaluate growth opportunities to further efforts in combating the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | | |
Collapse
|
45
|
De Plano LM, Saitta A, Oddo S, Caccamo A. Navigating Alzheimer's Disease Mouse Models: Age-Related Pathology and Cognitive Deficits. Biomolecules 2024; 14:1405. [PMID: 39595581 PMCID: PMC11592094 DOI: 10.3390/biom14111405] [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: 09/27/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Since the mid-1990s, scientists have been generating mouse models of Alzheimer's disease to elucidate key mechanisms underlying the onset and progression of the disease and aid in developing potential therapeutic approaches. The first successful mouse model of Alzheimer's disease was reported in 1995 with the generation of the PDAPP mice, which were obtained by the overexpression of gene coding for the amyloid precursor protein (APP). Since then, scientists have used different approaches to develop other APP overexpression mice, mice overexpressing tau, or a combination of them. More recently, Saito and colleagues generated a mouse model by knocking in mutations associated with familial Alzheimer's disease into the APP gene. In this review, we will describe the most used animal models and provide a practical guide for the disease's age of onset and progression. We believe that this guide will be valuable for the planning and experimental design of studies utilizing these mouse models.
Collapse
Affiliation(s)
| | | | | | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (L.M.D.P.); (A.S.); (S.O.)
| |
Collapse
|
46
|
Elariny HA, Kabel AM, Selim HMRM, Helal AI, Abdelrahman D, Borg HM, Elkady MA, Dawood LM, El-Badawy MF, Almalawi HFA, Arafa ESA, Alsufyani SE, Arab HH. Repositioning Canagliflozin for Mitigation of Aluminium Chloride-Induced Alzheimer's Disease: Involvement of TXNIP/NLRP3 Inflammasome Axis, Mitochondrial Dysfunction, and SIRT1/HMGB1 Signalling. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1805. [PMID: 39596990 PMCID: PMC11596744 DOI: 10.3390/medicina60111805] [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] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: Alzheimer's disease (AD) is the most common neurodegenerative disorder in the world. Due to failure of the traditional drugs to produce a complete cure for AD, the search for new safe and effective lines of therapy has attracted the attention of ongoing research. Canagliflozin is an anti-diabetic agent with proven efficacy in the treatment of neurological disorders in which mitochondrial dysfunction, oxidative stress, apoptosis, and autophagy play a pathophysiological role. Elucidation of the potential effects of different doses of canagliflozin on AD induced by aluminium chloride in rats and exploration of the molecular mechanisms that may contribute to these effects were the primary objectives of the current study. Materials and Methods: In a rat model of AD, the effect of three different doses of canagliflozin on the behavioural, biochemical, and histopathological alterations induced by aluminium chloride was assessed. Results: Canagliflozin administered to aluminium chloride-treated animals induced dose-dependent normalisation in the behavioural tests, augmentation of the antioxidant defence mechanisms, inhibition of TXNIP/NLRP3 inflammasome signalling, modulation of the SIRT1/HMGB1 axis, interference with the pro-inflammatory and the pro-apoptotic mechanisms, and restoration of the mitochondrial functions and autophagy in the hippocampal tissues to approximately baseline values. In addition, canagliflozin exhibited an interesting dose-dependent ability to repress aluminium chloride-induced histopathological changes in the brain. Conclusions: The effects of canagliflozin on oxidative stress, mitochondrial functions, inflammatory pathways, and autophagy signals may open new gates towards the mitigation of the pathologic features of AD.
Collapse
Affiliation(s)
- Hemat A. Elariny
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 55476, Saudi Arabia;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 35527, Egypt
| | - Ahmed M. Kabel
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.M.K.); (M.A.E.)
| | - Heba Mohammed Refat M. Selim
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia;
| | - Azza I. Helal
- Department of Histology and Cell Biology, Faculty of Medicine, Kafrelsheikh University, Kafr El-Shaikh 33516, Egypt;
| | - Doaa Abdelrahman
- Internal Medicine Department, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Hany M. Borg
- Physiology Department, Faculty of Medicine, Kafrelsheikh University, Kafr El-Shaikh 33516, Egypt;
| | - Mennatallah A. Elkady
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; (A.M.K.); (M.A.E.)
| | - Lamees M. Dawood
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt;
| | - Mohamed F. El-Badawy
- Microbiology and Immunology Department, Faculty of Pharmacy, University of Sadat City, Sadat City 32897, Egypt;
| | | | - El-Shaimaa A. Arafa
- College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates;
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Shuruq E. Alsufyani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (S.E.A.); (H.H.A.)
| | - Hany H. Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (S.E.A.); (H.H.A.)
| |
Collapse
|
47
|
Liu Z, Hu B, Tang J, Liu X, Cheng B, Jia C, Zhang L. Frontiers and hotspots evolution between air pollution and Alzheimer's disease: A bibliometric analysis from 2013 to 2023. J Alzheimers Dis 2024; 102:257-274. [PMID: 39573870 DOI: 10.1177/13872877241289381] [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: 11/26/2024]
Abstract
In recent years, the study of air pollution has received increasing attention from researchers, but a summary of Alzheimer's disease (AD) and air pollution is missed. Through combing the documents in the core dataset of Web of Science, this study analyzes current research based on specific keywords. CiteSpace and VOSviewer perform statistical analysis of measurement metrics to visualize a network of relevant content elements. The research devotes discussion to the relationship between air pollution and AD. Keyword hotspots include AD, children, oxidative stress, and system inflammation. Overall, 304 documents on air pollution and AD from 2013 to 2023 were retrieved from Web of Science. One hundred twenty-two journals published relevant articles, and the number of articles has increased gradually since the past decade. Research and development in AD and air pollution are progressing rapidly, but there is still a need for more connections with multidisciplinary technologies to explore cutting-edge hotspots.
Collapse
Affiliation(s)
- Zhirong Liu
- Department of General Surgery, The Affiliated Hospital of Chengdu Medical College, Chengdu Second People's Hospital, Chengdu, China
| | - BingShuang Hu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Ju Tang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - XinLian Liu
- Development and Regeneration Key Laboratory of Sichuan Province, Institute of Neuroscience, Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China
| | - BaoJing Cheng
- President Office, Chengdu Medical College, Chengdu, China
| | - Cui Jia
- Development and Regeneration Key Laboratory of Sichuan Province, Institute of Neuroscience, Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China
| | - LuShun Zhang
- Development and Regeneration Key Laboratory of Sichuan Province, Institute of Neuroscience, Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
48
|
Ren H, Tang L, Yuan Z, Liu Y, Zhou X, Xiao X, Wu X, Chen W, Chen Y, Wang H, Xue Q, Xu X. Combined administration of catalpol, puerarin, gastrodin, and borneol modulates the Tlr4/Myd88/NF-κB signaling pathway and alleviates microglia inflammation in Alzheimer's disease. Front Pharmacol 2024; 15:1492237. [PMID: 39545064 PMCID: PMC11560463 DOI: 10.3389/fphar.2024.1492237] [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: 09/10/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder affecting millions of people worldwide, with no effective treatment currently available. In recent decades, various traditional Chinese medicines (TCMs) and their active ingredients have shown the potential to attenuate the pathogenesis of AD in cellular and animal models. However, the effects of TCM formulas, which are typically administered in practice, have been less studied. This study aims to investigate the therapeutic effects of several formulas consisting of 4 components herbal components: catalpol, puerarin, gastrodin, and borneol, on streptozotocin (STZ)-induced AD models in cells and rats. The new object recognition (NOR), elevated plus maze (EMP), and Morris water maze (MWM) tests were used to evaluate the cognitive functions of rats. Golgi staining, Haematoxylin and Eosin (HE) staining, and Nissl staining analyses were employed assess the physiology of hippocampal tissues. Gene expression profiles were analyzed used transcriptomics and reverse transcription quantitative polymerase chain reaction analysis, while protein expression levels were determined using immunoblotting, immunohistochemical, and immunofluorescence. The production of cytokines was evaluated with enzyme-linked immunosorbent assay. The results demonstrated that the combined administration of these 4 components (CPGB) had superior mitigating effects on AD cell model, as evidenced by the reduced pro-inflammatory cytokine production and decreased deposition of Aβ protein. Further in vivo and in vitro experiments confirmed that varying doses of CPGB formula effectively ameliorated STZ-induced cognitive deficits, as shown by NOR, MWM, and EMP tests, as well as pathological changes in hippocampal tissues and a 3-dimensional brain neurovascular unit (3D-NVU) model, including decreased deposition of Aβ protein and formation of plaques. Transcriptome sequencing and analysis identified 35 genes with significantly altered expression levels due to STZ and CPGB treatment in hippocampal tissues, which were enriched in the Tlr4/Myd88/NF-κB signaling pathway. Interference with this pathway significantly influenced the therapeutic effects of CPGB in the 3D-NVU model. Collectively, these findings suggest that the combined administration of catalpol, puerarin, gastrodin, and borneol offers superior therapeutic effects on AD by modulating the Tlr4/Myd88/NF-κB signaling pathway. This study strengthens the theoretical foundation for using TCMs to treat AD, proving new insights and references for alleviating and treating AD.
Collapse
Affiliation(s)
- Huijing Ren
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- Tongren Polytechnic College, Tongren, Guizhou, China
| | - Ling Tang
- Shapingba District People’s Hospital of Chongqing, Chongqing, China
| | - Zhiying Yuan
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Yang Liu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xuejiao Zhou
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xiao Xiao
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xingyu Wu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Weihai Chen
- Faculty of Psychology, Southwest University, Chongqing, China
| | - Yi Chen
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Hongjin Wang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Qiang Xue
- Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Xiaoyu Xu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- Southwest University Hospital, Chongqing, China
- Chongqing Key Laboratory of New Drug Screening form Traditional Chinese Medicine, Chongqing, China
- Key Disciplines of Traditional Chinese Medicine of Chongqing City, Rehabilitation Medicine of Southwest University, Chongqing, China
| |
Collapse
|
49
|
Birnbaum EM, Xie L, Serrano P, Rockwell P, Figueiredo-Pereira ME. BT-11 repurposing potential for Alzheimer's disease and insights into its mode of actions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620882. [PMID: 39553925 PMCID: PMC11565763 DOI: 10.1101/2024.10.29.620882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Neuroinflammation is a key pathological hallmark of Alzheimer's disease (AD). Investigational and FDA approved drugs targeting inflammation already exist, thus drug repurposing for AD is a suitable approach. BT-11 is an investigational drug that reduces inflammation in the gut and improves cognitive function. BT-11 is orally active and binds to lanthionine synthetase C-like 2 (LANCL2), a glutathione-s-transferase, thus potentially reducing oxidative stress. We investigated the effects of BT-11 long-term treatment on the TgF344-AD rat model. BT-11 reduced hippocampal-dependent spatial memory deficits, Aβ plaque load and neuronal loss in males, and mitigated microglia numbers in females. BT-11 treatment led to hippocampal transcriptomic changes in signaling receptor, including G-protein coupled receptor pathways. We detected LANCL2 in hippocampal nuclear and cytoplasmic fractions with potential different post-translational modifications, suggesting distinct functions based on its subcellular localization. LANCL2 was present in oligodendrocytes, showing a role in oligodendrocyte function. To our knowledge, these last two findings have not been reported. Overall, our data suggest that targeting LANCL2 with BT-11 improves cognition and reduces AD-like pathology by potentially modulating G-protein signaling and oligodendrocyte function. Our studies contribute to the field of novel immunomodulatory AD therapeutics, and merit further research on the role of LANCL2 in this disease.
Collapse
|
50
|
Golipoor M, Rafat Z, Saberi A, Roostaei D, Shabanpour AM. Comparing the frequency, antifungal susceptibility, and enzymatic profiles of the oral fungal composition in patients with and without Alzheimer's disease admitted to a neurology clinic. Front Cell Infect Microbiol 2024; 14:1477230. [PMID: 39492992 PMCID: PMC11527782 DOI: 10.3389/fcimb.2024.1477230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Background Studies have shown that changes in the frequency of oral microorganisms may play a key role in the development of Alzheimer's disease (AD). However, no research has been conducted on the oral fungal composition in AD-patients. The present study aimed to investigate the changes in the frequency of oral fungal composition, the antifungal susceptibility, and the enzymatic profiles of oral fungal composition in patients suffering from AD compared to non-AD individuals. Materials and methods In the present analytical cross-sectional study during 12 months, 76 hospitalized patients with AD were matched with 76 individuals without AD. A sterile serum physiology-moistened cotton-tipped swab was used to sample the mouth area. All swabs were cultured on Sabouraud Chloramphenicol Agar. Fungal identified were confirmed through the PCR-sequencing techniques. Enzyme activity index (EAI) for important pathogenic factors including proteinase, esterase and hemolysin was measured using relevant protocols. The susceptibility to 8 antifungal agents (nystatin, voriconazole, itraconazole, fluconazole, posaconazole, amphotericin B, 5-fluorocytosine, and caspofungin) against fungal strains obtained from AD-patients was evaluated according to the Clinical and Laboratory Standards Institute (CLSI) guidelines, document M38-A2 for filamentous fungi, and document M27-A4 for yeasts. Results The results showed that compared to the non-AD individuals, the prevalence of oral fungal composition in AD group was 1.6 times higher. Candida albicans was the most common fungal species isolated from oral swab samples of AD group (n=53, 80%) and non-AD group (n=28, 40%), and the diversity of the oral fungal composition in AD-patients were lower than non-AD individuals. Among the 3 investigated virulence factors, a statistically significant difference was shown in terms of hemolysin activity level between the two studied groups (p<0.05) and the activity level of esterase and proteinase enzymes did not show a significant difference in the two studied groups (p>0.05). The results showed that almost all of the tested isolates were susceptible to nystatin, the most widely prescribed antifungal to treat superficial infections, and only 1.69 % (2/118) of the Candida isolates were resistant to this antifungal drug. Conclusion Understanding the changes in the frequency of oral fungal composition the antifungal susceptibility, and the enzymatic profiles of oral fungal composition in patients suffering from AD compared to non-AD individuals makes it possible to better understand the etiology of this disease.
Collapse
Affiliation(s)
- Mandana Golipoor
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Rafat
- Department of Medical Parasitology and Mycology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Alia Saberi
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Davoud Roostaei
- Department of Pharmacology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amir-Mohammad Shabanpour
- Student research committee, Anzali International Medical Campus, Guilan University of Medical Sciences, Guilan, Iran
| |
Collapse
|