1
|
Fu M, Wang QW, Liu YR, Chen SJ. The role of the three major intestinal barriers in ulcerative colitis in the elderly. Ageing Res Rev 2025; 108:102752. [PMID: 40210198 DOI: 10.1016/j.arr.2025.102752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/21/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
With the unprecedented pace of global population aging, there has been a parallel epidemiological shift marked by increasing incidence rates of ulcerative colitis (UC) in geriatric populations, imposing a substantial disease burden on healthcare systems globally. The etiopathogenesis of UC in the elderly remains poorly delineated, while current therapeutic strategies require further optimization to accommodate the unique pathophysiological characteristics of elderly patients. This review systematically elucidates the three barrier dysfunction - encompassing the gut microbiota ecosystem, mucosal epithelial integrity, and immunoregulatory network - that collectively drives UC pathogenesis during biological senescence. We emphasize the therapeutic potential of barrier-targeted interventions, particularly highlighting emerging modalities including fecal microbiota transplantation, intestinal organoid regeneration techniques, mesenchymal stem cell-mediated immunomodulation, and precision-engineered Chimeric Antigen Receptor T-cell therapies. Through this multidimensional analysis, we propose a paradigm-shifting approach to UC management in the elderly, advocating for the development of tailored and evidence-based therapeutic interventions that address the complex interplay between age-related biological changes and intestinal barrier homeostasis in elderly patients.
Collapse
Affiliation(s)
- Min Fu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Qi-Wen Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Ya-Ru Liu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Shu-Jie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China.
| |
Collapse
|
2
|
Assaifan AK, Alfadul H, Albuaimi MS, Alrebaish AS, Al-Gawati M. Scalable flexographic printing of graphite/carbon dot nanobiosensors for non-faradaic electrochemical quantification of IL-8. Talanta 2025; 295:128371. [PMID: 40412198 DOI: 10.1016/j.talanta.2025.128371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/28/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Interleukin-8 (IL-8) is a key biomarker linked to inflammation and disability in neonates. However, current IL-8 detection methods are often costly, labor-intensive, and require highly trained personnel. While electrochemical techniques have been employed for sensitive IL-8 quantification, they typically rely on redox probes and three-electrode electrochemical cells, leading to issues such as toxicity, prolonged fabrication time, and increased waste generation. Additionally, conventional electrochemical biosensors fabrication techniques are expensive and time-consuming, limiting their scalability for mass disease screening. In this study, we introduce a low-cost, non-faradaic electrochemical nanobiosensor for the direct detection of IL-8. The sensor consists of interdigitated graphite/carbon dot conjugates flexographically printed onto a flexible polyimide substrate. The printed layer's physical properties were systematically characterized using SEM, AFM and surface profilometer, and biofunctionalization was achieved using aminopropyltriethoxysilane (APTES) and glutaraldehyde. Successful surface modification was confirmed through ATR-FTIR and EDS elemental mapping. Electrochemical impedance spectroscopy (EIS) analysis demonstrated the nanobiosensor's response to varying IL-8 concentrations, with capacitance, Zmod, Zreal, and Zimag measurements. Among these, Zimag exhibited the highest sensitivity, with a response of 2.7 kΩ/log(ng/mL) and a detection limit of 50 pg/mL-well below the clinically established threshold of 600 pg/mL. This study demonstrates that, in addition to capacitance, there are multiple parameters that warrant exploration in non-faradaic biosensors to improve their sensing performance. The nanobiosensor fabrication via flexographic printing enables scalable, cost-effective production while maintaining high sensitivity and selectivity through a non-faradaic detection mechanism. This work paves the way for the development of affordable, mass-producible biosensors for early biomarker detection, facilitating timely medical intervention and improved neonatal healthcare outcomes.
Collapse
Affiliation(s)
- Abdulaziz K Assaifan
- Department of Biomedical Technology, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia; King Salman Center for Disability Research, Riyadh, 11614, Saudi Arabia; Biological and Environmental Sensing Research Unit, King Abdullah Institute for Nanotechnology, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.
| | - Hend Alfadul
- Department of Biomedical Technology, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia; Biological and Environmental Sensing Research Unit, King Abdullah Institute for Nanotechnology, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Munira S Albuaimi
- Department of Biomedical Technology, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia; Biological and Environmental Sensing Research Unit, King Abdullah Institute for Nanotechnology, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Abdulelah S Alrebaish
- Department of Biomedical Technology, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Mahmoud Al-Gawati
- Biological and Environmental Sensing Research Unit, King Abdullah Institute for Nanotechnology, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
3
|
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
|
4
|
GuoYan L, ShuQin Z, HuiYang Q, MengYing C, HongWei R, Hong S. IL-8 and IL-1β in the Cerebrospinal Fluid of Patient With Creutzfeldt-Jakob Disease: A Case Report. Clin Case Rep 2025; 13:e70508. [PMID: 40370789 PMCID: PMC12076592 DOI: 10.1002/ccr3.70508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/17/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025] Open
Abstract
Inflammatory cytokines play a crucial role in Creutzfeldt-jakob disease (CJD). We examined inflammatory cytokine levels in both blood and CSF in a CJD patient and an epileptic encephalopathy as a control case. Results showed that IL-6, IL-8, IL-1β, and TNF-α levels were elevated in blood, but only IL-8 and IL-1β levels, especially IL-8, were significantly elevated in Cerebrospinal fluid (CSF). This suggests that IL-8 and IL-1β are closely related to cognitive impairment and blood-brain barrier (BBB) damage in CJD, and IL-8 might be a key target in early cognitive impairment in CJD.
Collapse
Affiliation(s)
- Liu GuoYan
- Department of NeurologyThe Second Affiliated Hospital, Medical School of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Zhan ShuQin
- Department of NeurologyThe Second Affiliated Hospital, Medical School of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Qu HuiYang
- Department of NeurologyThe Second Affiliated Hospital, Medical School of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Chen MengYing
- Department of NeurologyThe Second Affiliated Hospital, Medical School of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Ren HongWei
- Department of NeurologyThe Second Affiliated Hospital, Medical School of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Sun Hong
- Department of NeurologyThe Second Affiliated Hospital, Medical School of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
5
|
Chagas YW, Vaz de Castro PAS, Simões-E-Silva AC. Neuroinflammation in kidney disease and dialysis. Behav Brain Res 2025; 483:115465. [PMID: 39922385 DOI: 10.1016/j.bbr.2025.115465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/10/2025]
Abstract
The complex relationship between chronic kidney disease (CKD) and neuroinflammation shows how important immunological processes are in mediating cognitive dysfunction and psychiatric symptoms in this disease. Proinflammatory cytokines and chemokines, such as IL-1β and IL-6, are capable of crossing the blood-brain barrier, and, consequently, may contribute to neuropsychiatric symptoms including anxiety, depression, and cognitive impairment in CKD patients. The peptides of the renin-angiotensin system (RAS), with their dual functions in inflammation and neuroprotection, also highlight the intricate immunological mechanisms operating within the kidney-brain axis. Understanding these immunological pathways is essential for developing targeted interventions to modulate neuroinflammation and improve cognitive outcomes in individuals with CKD. Further research in renal immunology and neuroinflammation holds promise for advancing our understanding of the intricate connections between kidney health, brain function, and immune responses in the context of CKD. This review summarizes the critical role of immunological factors in the pathophysiology of CKD-related cognitive impairment and psychiatric disorders.
Collapse
Affiliation(s)
- Yumi Watanabe Chagas
- Interdisciplinary Laboratory of Medical Investigation, Unit of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Brazil
| | - Pedro Alves S Vaz de Castro
- Interdisciplinary Laboratory of Medical Investigation, Unit of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Brazil
| | - Ana Cristina Simões-E-Silva
- Interdisciplinary Laboratory of Medical Investigation, Unit of Pediatric Nephrology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Brazil.
| |
Collapse
|
6
|
Capogna E, Sørensen Ø, Watne LO, Roe J, Strømstad M, Idland AV, Halaas NB, Blennow K, Zetterberg H, Walhovd KB, Fjell AM, Vidal-Piñeiro D. Subtypes of brain change in aging and their associations with cognition and Alzheimer's disease biomarkers. Neurobiol Aging 2025; 147:124-140. [PMID: 39740372 DOI: 10.1016/j.neurobiolaging.2024.12.009] [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: 03/22/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
Structural brain changes underlie cognitive changes and interindividual variability in cognition in older age. By using structural MRI data-driven clustering, we aimed to identify subgroups of cognitively unimpaired older adults based on brain change patterns and assess how changes in cortical thickness, surface area, and subcortical volume relate to cognitive change. We tested (1) which brain structural changes predict cognitive change (2) whether these are associated with core cerebrospinal fluid (CSF) Alzheimer's disease biomarkers, and (3) the degree of overlap between clusters derived from different structural modalities in 1899 cognitively healthy older adults followed up to 16 years. We identified four groups for each brain feature, based on the degree of a main longitudinal component of decline. The minimal overlap between features suggested that each contributed uniquely and independently to structural brain changes in aging. Cognitive change and baseline cognition were associated with cortical area change, whereas higher baseline levels of phosphorylated tau and amyloid-β related to changes in subcortical volume. These results may contribute to a better understanding of different aging trajectories.
Collapse
Affiliation(s)
- Elettra Capogna
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo 0373, Norway.
| | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo 0373, Norway
| | - Leiv Otto Watne
- Department of Geriatric Medicine, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - James Roe
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo 0373, Norway
| | - Marie Strømstad
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo 0373, Norway
| | - Ane Victoria Idland
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Nathalie Bodd Halaas
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, Campus Ullevål, University of Oslo, Oslo, Norway.
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, PR China
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Center for Neurodegenerative Diseases, Hong Kong; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kristine Beate Walhovd
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo 0373, Norway; Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Anders Martin Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo 0373, Norway; Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Didac Vidal-Piñeiro
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo 0373, Norway
| |
Collapse
|
7
|
Jiao W, Lin J, Deng Y, Ji Y, Liang C, Wei S, Jing X, Yan F. The immunological perspective of major depressive disorder: unveiling the interactions between central and peripheral immune mechanisms. J Neuroinflammation 2025; 22:10. [PMID: 39828676 PMCID: PMC11743025 DOI: 10.1186/s12974-024-03312-3] [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: 09/14/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025] Open
Abstract
Major depressive disorder is a prevalent mental disorder, yet its pathogenesis remains poorly understood. Accumulating evidence implicates dysregulated immune mechanisms as key contributors to depressive disorders. This review elucidates the complex interplay between peripheral and central immune components underlying depressive disorder pathology. Peripherally, systemic inflammation, gut immune dysregulation, and immune dysfunction in organs including gut, liver, spleen and adipose tissue influence brain function through neural and molecular pathways. Within the central nervous system, aberrant microglial and astrocytes activation, cytokine imbalances, and compromised blood-brain barrier integrity propagate neuroinflammation, disrupting neurotransmission, impairing neuroplasticity, and promoting neuronal injury. The crosstalk between peripheral and central immunity creates a vicious cycle exacerbating depressive neuropathology. Unraveling these multifaceted immune-mediated mechanisms provides insights into major depressive disorder's pathogenic basis and potential biomarkers and targets. Modulating both peripheral and central immune responses represent a promising multidimensional therapeutic strategy.
Collapse
Affiliation(s)
- Wenli Jiao
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Jiayi Lin
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Yanfang Deng
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yelin Ji
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Chuoyi Liang
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Sijia Wei
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China
| | - Xi Jing
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geoscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, Guangdong, China.
| | - Fengxia Yan
- School of Nursing, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|
8
|
Bruno M, Bonomi CG, Castelli A, Izzi F, Placidi F, Falletti S, Mercuri NB, Motta C, Martorana A. Cerebrospinal fluid cytokine levels affect electroencephalographic activity in Alzheimer's disease. J Alzheimers Dis Rep 2025; 9:25424823241306772. [PMID: 40165839 PMCID: PMC11956505 DOI: 10.1177/25424823241306772] [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: 05/31/2024] [Accepted: 11/11/2024] [Indexed: 04/02/2025] Open
Abstract
To investigate the role of neuroinflammation as mediator of amyloid-β-induced cortical activity changes in Alzheimer's disease (AD), we examined the relationship between cerebrospinal fluid (CSF) inflammatory cytokines (IL-1β, IL-2, IL-4, IL-6, IL-7, IL-8, IL-10, IL-12, IL-13, IL-17, TNF-α, IFN-γ, GM-CSF, G-CSF, MIP-1α, MCP-1) and electroencephalographic (EEG) abnormalities in a cohort of biologically defined AD patients (n = 55, M:F = 19:36, median age 73, Mini-Mental State Examination ≥ 22). We retrieved a positive association between IL-4 CSF levels and EEG background activity frequency; IL-7, IL-8, and IL-12 CSF levels were positively associated with the presence of interictal epileptiform discharges. Neuroinflammation accompanying AD pathology may enhance the amyloid's epileptogenic potential while also counteracting neurodegenerative damage.
Collapse
Affiliation(s)
- Matilde Bruno
- Memory Clinic, Policlinico Tor Vergata, University of Rome “Tor Vergata” – Rome, Italy
| | | | - Alessandro Castelli
- Epilepsy Centre, Policlinico Tor Vergata, University of Rome Tor Vergata – Rome, Italy
| | - Francesca Izzi
- Epilepsy Centre, Policlinico Tor Vergata, University of Rome Tor Vergata – Rome, Italy
| | - Fabio Placidi
- Epilepsy Centre, Policlinico Tor Vergata, University of Rome Tor Vergata – Rome, Italy
| | - Silvia Falletti
- Memory Clinic, Policlinico Tor Vergata, University of Rome “Tor Vergata” – Rome, Italy
| | | | - Caterina Motta
- Memory Clinic, Policlinico Tor Vergata, University of Rome “Tor Vergata” – Rome, Italy
| | - Alessandro Martorana
- Memory Clinic, Policlinico Tor Vergata, University of Rome “Tor Vergata” – Rome, Italy
| |
Collapse
|
9
|
Righi D, Manco C, Pardini M, Stufano A, Schino V, Pelagotti V, Massa F, Stefano ND, Plantone D. Investigating interleukin-8 in Alzheimer's disease: A comprehensive review. J Alzheimers Dis 2025; 103:38-55. [PMID: 39558604 DOI: 10.1177/13872877241298973] [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: 11/20/2024]
Abstract
Several studies indicate that the development of Alzheimer's disease (AD) has strong interactions with immune mechanisms within the brain, indicating a close association between inflammation in the central nervous system and the progression of neurodegeneration. Despite considerable progress in understanding the inflammatory aspects of AD, several of them remain unresolved. Pro-inflammatory cytokines and microglia are pivotal components in the inflammatory cascade. Among these, the role of interleukin-8 (IL-8) in neurodegeneration seems complex and multifaceted, involving inflammation, neurotoxicity, blood-brain barrier disruption, and oxidative stress, and is still poorly characterized. We conducted a review to describe the evidence of IL-8 involvement in AD. IL-8 is a cytokine known for its proinflammatory properties and typically produced by macrophages, predominantly functions as a chemotactic signal for attracting neutrophils to inflamed sites in the bloodstream. Interestingly, IL-8 is also present in the brain, where it is primarily released by microglia in response to inflammatory signals. This review aims to provide a comprehensive overview of the structure, function, and regulatory mechanisms of IL-8 relevant to AD pathology.
Collapse
Affiliation(s)
- Delia Righi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Carlo Manco
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Angela Stufano
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Valentina Schino
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Virginia Pelagotti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Domenico Plantone
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
10
|
Grigore M, Ruscu MA, Hermann DM, Colita IC, Doeppner TR, Glavan D, Popa-Wagner A. Biomarkers of cognitive and memory decline in psychotropic drug users. J Neural Transm (Vienna) 2025; 132:39-59. [PMID: 39377784 PMCID: PMC11735527 DOI: 10.1007/s00702-024-02837-4] [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: 08/17/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024]
Abstract
Psychotropic drugs are vital in psychiatry, aiding in the management of mental health disorders. Their use requires an understanding of their pharmacological properties, therapeutic applications, and potential side effects. Ongoing research aims to improve their efficacy and safety. Biomarkers play a crucial role in understanding and predicting memory decline in psychotropic drug users. A comprehensive understanding of biomarkers, including neuroimaging, biochemical, genetic, and cognitive assessments, is essential for developing targeted interventions and preventive strategies. In this narrative review, we performed a comprehensive search on PubMed and Google using review-specific terms. Clinicians should use a multifaceted approach, including neurotransmitter analysis, neurotrophic factors, miRNA profiling, and cognitive tasks for early intervention and personalized treatment. Anxiolytics' mechanisms involve various neurotransmitter systems and emerging targets. Research on biomarkers for memory decline in anxiolytic users can lead to early detection and intervention, enhancing clinical practices and aligning with precision medicine. Mood stabilizer users can benefit from early detection of memory decline through RNA, neurophysiological, and inflammatory biomarkers, promoting timely interventions. Performance-enhancing drugs may boost athletic performance in the short term, but their long-term health risks and ethical issues make their use problematic. Long-term use of psychotropic performance enhancers in athletes shows changes in biomarkers of cognitive decline, necessitating ongoing monitoring and intervention strategies. Understanding these genetic influences on memory decline helps pave the way for personalized approaches to prevent or mitigate cognitive deterioration, emphasizing the importance of genetic screening and early interventions based on an individual's genetic profile. Future research should focus on refining these biomarkers and protective measures against cognitive deterioration. Overall, a comprehensive understanding of biomarkers in psychotropic drug users is essential for developing targeted interventions and preventive strategies.
Collapse
Affiliation(s)
- Monica Grigore
- Department of Psychiatry, University of Medicine and Pharmacy Craiova, Petru Rares 2-4, 200349, Romania, Craiova
| | - Mihai Andrei Ruscu
- Doctoral School, University of Medicine and Pharmacy Craiova, 200349, Craiova, Romania
| | - Dirk M Hermann
- Chair of Vascular Neurology, Dementia and Ageing, University Hospital Essen, 45147, Essen, Germany
- Department of Psychiatry, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | - Ivan-Cezar Colita
- Department of Neurology, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Thorsten Roland Doeppner
- Department of Neurology, University Medical Center Göttingen, 37075, Göttingen, Germany
- Department of Neurology, University of Giessen Medical School, 35392, Giessen, Germany
| | - Daniela Glavan
- Department of Psychiatry, University of Medicine and Pharmacy Craiova, Petru Rares 2-4, 200349, Romania, Craiova.
| | - Aurel Popa-Wagner
- Chair of Vascular Neurology, Dementia and Ageing, University Hospital Essen, 45147, Essen, Germany.
| |
Collapse
|
11
|
Ormiston K, Melink Z, Andridge R, Lustberg M, Courtney DeVries A, Murphy K, Emmers K, Ziouzenkova O, Belury MA, Orchard TS. Dietary EPA and DHA enrichment of a high fat diet during doxorubicin-based chemotherapy attenuated neuroinflammatory gene expression in the brain of C57bl/6 ovariectomized mice. Brain Behav Immun 2025; 123:370-382. [PMID: 39313165 DOI: 10.1016/j.bbi.2024.09.021] [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: 05/20/2024] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
Chemotherapy agents in breast cancer are associated with chemotherapy-related cognitive impairments (CRCI). Mechanisms are not fully clear, but alterations of glucose and lipid metabolism, neuroinflammation and neurodegeneration may contribute to CRCI. The aim of this study was to investigate the combined effects of a high fat (HF) diet combined with doxorubicin-based chemotherapy on glucose and lipid metabolism, neuroinflammation, and neurodegeneration in mice. Additionally, we examined the therapeutic potential of dietary eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) to attenuate these effects. Female C57Bl/6 mice (n = 42) were fed HF, HFn-3 (2 % kcals as EPA + DHA) or Low Fat (LF) diets for seven weeks, with and without chemotherapy. In this study, two chemotherapy injections led to weight and body fat loss associated with a decrease in insulin resistance measured by HOMA-IR. HOMA-IR was significantly greater in HF versus LF groups; but HOMA-IR in HFn-3 group did not significantly differ from either HF or LF groups. Chemotherapy resulted in higher brain concentrations of the inflammatory chemokine KC/GRO. Compared to LF diet plus chemotherapy, HF diet plus chemotherapy upregulated multiple genes involved in neuroinflammation and neurodegeneration pathways. HFn-3 diet plus chemotherapy attenuated gene expression by downregulating multiple genes involved in neuroinflammation and blood brain barrier regulation, including Mapkapk2, Aqp4, and s100b, and upregulating Kcnb1 and Atxn3, genes involved in reduction of oxidative stress and anxiety, respectively. Overall, a HF diet combined with chemotherapy is associated with neuroinflammatory and neurodegenerative gene expression changes in this mouse model; dietary enrichment of EPA and DHA attenuated these effects. Further studies are needed to understand how diet impacts behavioral outcomes of CRCI.
Collapse
Affiliation(s)
- Kate Ormiston
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Zihan Melink
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Rebecca Andridge
- Division of Biostatistics, College of Public Health, The Ohio State University, United States
| | | | | | - Kelly Murphy
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Katie Emmers
- Department of Veterinary Medicine, The Ohio State University, United States
| | - Ouliana Ziouzenkova
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Martha A Belury
- Department of Food Science and Technology, The Ohio State University, United States
| | - Tonya S Orchard
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States.
| |
Collapse
|
12
|
Jin D, Zhang M, Shi L, Liu H. Investigating the Impact of IL-6 and CXCL8 on Neurodegeneration and Cognitive Decline in Alzheimer Disease. Int J Neuropsychopharmacol 2024; 28:pyae038. [PMID: 39223908 PMCID: PMC11781222 DOI: 10.1093/ijnp/pyae038] [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: 05/01/2024] [Accepted: 09/02/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Alzheimer disease (AD) is a progressive neurodegenerative disorder primarily affecting the elderly, characterized by severe cognitive impairment and memory loss. Emerging evidence suggests that neuroinflammation plays a significant role in AD pathogenesis, with cytokines like interleukin-6 (IL-6) and C-X-C motif chemokine ligand 8 (CXCL8) contributing to the disease progression. METHODS We utilized Gene Expression Omnibus datasets to identify IL-6 and CXCL8 as pivotal inflammatory markers in AD. In vitro experiments were conducted using SK-N-BE(2)-M17 and THP-1 cell lines treated with IL-6 and CXCL8 to model AD. Additionally, in vivo tests on Amyloid Precursor Protein/Presenilin 1 (APP/PS1) AD mouse models were performed to assess the impact of these cytokines on cognitive functions and brain pathology. RESULTS The results indicated a significant decrease in cell viability, increased apoptosis, and elevated inflammatory factor secretion following IL-6 and CXCL8 treatment in vitro. In vivo, AD mouse models treated with these cytokines exhibited exacerbated emotional distress, decreased social interaction, impaired cognitive functions, and increased amyloid protein deposition in neural tissues. CONCLUSIONS The study highlights the detrimental effects of IL-6 and CXCL8 on neuronal health and cognitive functions in AD. These findings suggest that targeting these cytokines could offer potential therapeutic interventions for improving patient outcomes in Alzheimer disease.
Collapse
Affiliation(s)
- Dongdong Jin
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Min Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Shi
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hengfang Liu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Melink Z, Lustberg MB, Schnell PM, Mezzanotte-Sharpe J, Orchard TS. Effect of minocycline on changes in affective behaviors, cognitive function, and inflammation in breast cancer survivors undergoing chemotherapy: a pilot randomized controlled trial. Breast Cancer Res Treat 2024; 208:605-617. [PMID: 39143391 PMCID: PMC11522141 DOI: 10.1007/s10549-024-07457-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: 02/20/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE Minocycline suppresses chemotherapy-induced neuroinflammation in preclinical models, but its effects in cancer survivors are unknown. This study evaluated the longitudinal effects of minocycline on affective behaviors, cognitive functions, and inflammation in women with breast cancer (BC) undergoing chemotherapy. METHODS This is a pilot, double-blind, randomized controlled trial of oral minocycline (100 mg BID) versus placebo for chemotherapy-induced affective disorders in women initiating chemotherapy for stage I-III BC. Participants received minocycline or placebo up to one week before chemotherapy, continuing through cycle 4 (C4). Epidemiologic Studies Depression Scale (CES-D) and State-Trait Anxiety Inventory (STAI) were assessed at baseline, each cycle of chemotherapy (C1-C4), 2-3-week post-chemotherapy (end of chemotherapy), and 6-month post-chemotherapy (6 M) as the primary outcomes. Sub-group analysis of CES-D and STAI based on the severity of symptoms was also performed. Changes in self-reported cognition and serum inflammatory markers were also evaluated. RESULTS Fifty-seven women enrolled and 55 completed the study. Except for Interleukin-8 (p ≤ 0.03), changes in inflammatory markers, cognitive function, CES-D, and STAI were not significantly different between groups from baseline to any cycle or post-chemotherapy time point (all p > 0.05), adjusting for baseline scores. Increases in serum Interleukin-8 from baseline to C4 and 6 M were ameliorated by minocycline (p < 0.05). The sub-group symptomatic for depression (CES-D > = 16 at baseline) treated with minocycline had a greater reduction in CES-D score compared to placebo from baseline to 6 M (p = 0.01). CONCLUSION Despite attenuation of IL-8, minocycline did not alter self-reported affective symptoms or cognition in this cohort of BC survivors undergoing chemotherapy. The effect of minocycline on BC survivors symptomatic for depression before chemotherapy warrants further investigation.
Collapse
Affiliation(s)
- Zihan Melink
- Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Maryam B Lustberg
- Yale School of Medicine, Center for Breast Cancer, New Haven, CT, 06511, USA
| | - Patrick M Schnell
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Jessica Mezzanotte-Sharpe
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Tonya S Orchard
- Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
14
|
Chen J, Ragab AAY, Doyle MF, Alosco ML, Fang Y, Mez J, Satizabal CL, Qiu WQ, Murabito JM, Lunetta KL. Inflammatory protein associations with brain MRI measures: Framingham Offspring Cohort. Alzheimers Dement 2024; 20:7465-7478. [PMID: 39282876 PMCID: PMC11567832 DOI: 10.1002/alz.14147] [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] [Revised: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Brain magnetic resonance imaging (MRI) and inflammatory biomarkers are crucial for investigating preclinical neurocognitive disorders. Current investigations focus on a few inflammatory markers. The study aims to investigate the associations between inflammatory biomarkers and MRI measures and to examine sex differences among the associations in the Framingham Heart Study. METHODS Dementia and stroke-free participants underwent OLINK Proteomics profiling and MRI measurements within 5 years. Pairwise cross-sectional analysis assessed 68 biomarkers with 13 brain MRI volumes, adjusting for covariates and familial correlations. RESULTS Elevated CDCP1, IL6, OPG, and 4E.BP1 were related to smaller total cerebral brain volume (TCBV), whereas higher HGF, IL8, and MMP10 were associated with smaller TCBV, total and frontal white matter volumes. Higher SCF and TWEAK were associated with larger TCBV. In sex-stratified analyses, associations were observed exclusively among males. DISCUSSION We report several associations between inflammatory biomarkers and brain volumes, highlighting different associations within sex subgroups. HIGHLIGHTS Higher CDCP1, IL6, OPG, and 4E.BP1 levels were associated with smaller TCBV. Higher levels of HGF, IL8 and MMP10 were associated with smaller TCBV, CWV and FWV. Higher levels of SCF and TWEAK, were associated with larger TCBV. Significance diminished in models adjusting for CVD risk factors. Associations were observed exclusively in males.
Collapse
Affiliation(s)
- Jiachen Chen
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Ahmed A. Y. Ragab
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Margaret F. Doyle
- Department of Pathology and Laboratory MedicineLarner College of MedicineUniversity of VermontBurlingtonVermontUSA
| | - Michael L. Alosco
- Boston University Chobanian & Avedisian School of MedicineBoston University Alzheimer's Disease Research Center and CTE CenterBostonMassachusettsUSA
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Yuan Fang
- School of Pharmacy and Pharmaceutical SciencesBinghamton UniversityState University of New YorkBinghamtonNew YorkUSA
| | - Jesse Mez
- Boston University Chobanian & Avedisian School of MedicineBoston University Alzheimer's Disease Research Center and CTE CenterBostonMassachusettsUSA
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University School of MedicineFraminghamMassachusettsUSA
| | - Claudia L. Satizabal
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUniversity of Texas Health Science Center at San AntonioSan AntonioTexasUSA
| | - Wei Qiao Qiu
- Boston University Chobanian & Avedisian School of MedicineBoston University Alzheimer's Disease Research Center and CTE CenterBostonMassachusettsUSA
- Department of PsychiatryBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of Pharmacology & Experimental TherapeuticsBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Joanne M. Murabito
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University School of MedicineFraminghamMassachusettsUSA
- Department of MedicineSection of General Internal MedicineBoston University Chobanian & Avedisian School of Medicine and Boston Medical CenterBostonMassachusettsUSA
| | - Kathryn L. Lunetta
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| |
Collapse
|
15
|
Long J, Zhang J, Zeng X, Wang M, Wang N. Prevention and Treatment of Alzheimer's Disease Via the Regulation of the Gut Microbiota With Traditional Chinese Medicine. CNS Neurosci Ther 2024; 30:e70101. [PMID: 39508315 PMCID: PMC11541599 DOI: 10.1111/cns.70101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
Alzheimer's disease (AD) is caused by a variety of factors, and one of the most important factors is gut microbiota dysbiosis. An imbalance in the gut mincrobiota have been shown to change the concentrations of lipopolysaccharide and short-chain fatty acids. These microorganisms synthesize substances that can influence the levels of a variety of metabolites and cause multiple diseases through the immune response, fatty acid metabolism, and amino acid metabolism pathways. Furthermore, these metabolic changes promote the formation of β-amyloid plaques and neurofibrillary tangles. Thus, the microbiota-gut-brain axis plays an important role in AD development. In addition to traditional therapeutic drugs such as donepezil and memantine, traditional Chinese medicines (TCMs) have also showed to significantly decrease the severity of AD symptoms and suppress the underlying related mechanisms. We searched for studies on the effects of different herbal monomers, single herbs, and polyherbal formulas on the gut microbiota of AD patients and identified the relevant pathways through which the gut microbiota affected AD. We conclude that improvements in the gut microbiota not only decrease the occurrence of inflammatory reactions but also reduce the deposition of central pathological products. Herbal monomers have a stronger effect on improving of central pathology. Polyherbal formulas have the most extensive effect on the gut microbiota in patients with AD. Among the effects of formulas, the anti-inflammatory effect is the most essential and is also the main concern regarding the use of TCMs in treating AD from the viewpoint of the gut microbiota. We hope that this review will be helpful for providing new ideas for the clinical application of TCMs in the treatment of AD.
Collapse
Affiliation(s)
- Jinyao Long
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Jiani Zhang
- School of Life SciencesBeijing University of Chinese MedicineBeijingChina
| | - Xin Zeng
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Min Wang
- Dongfang Hospital Beijing University of Chinese MedicineBeijingChina
| | - Ningqun Wang
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
16
|
Galluzzi S, Marizzoni M, Gatti E, Bonfiglio NS, Cattaneo A, Epifano F, Frisoni GB, Genovese S, Geviti A, Marchetti L, Sgrò G, Solorzano CS, Pievani M, Fiorito S. Citrus supplementation in subjective cognitive decline: results of a 36-week, randomized, placebo-controlled trial. Nutr J 2024; 23:135. [PMID: 39482712 PMCID: PMC11529263 DOI: 10.1186/s12937-024-01039-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Developing interventions for older adults with subjective cognitive decline (SCD) has the potential to prevent dementia in this at-risk group. Preclinical models indicate that Citrus-derived phytochemicals could benefit cognition and inflammatory processes, but results from clinical trials are still preliminary. The aim of this study is to determine the effects of long-term supplementation with Citrus peel extract on cognitive performance and inflammation in individuals with SCD. METHODS Eighty participants were randomly assigned to active treatment (400 mg of Citrus peel extract containing 3.0 mg of naringenin and 0.1 mg of auraptene) or placebo at 1:1 ratio for 36 weeks. The primary endpoint was the change in the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) total score across the 36-week trial period. Other cognitive outcomes included tests and scales evaluating verbal memory, attention, executive and visuospatial functions, and memory concerns. The secondary endpoint was the change of interleukin-8 (IL-8) levels over the 36-week trial period in a subsample of 60 consecutive participants. An Intention-to-treat approach with generalized linear mixed models was used for data analysis. RESULTS The RBANS total score showed significant improvement in both Citrus peel extract and placebo groups at 36 weeks (p for time < .001, d = 0.36, p time x treatment = .910). Significant time effects were also found in cognitive domains of short- and long-term verbal memory (p < .001) and scales of subjective memory (p < .01), with no significant time x treatment interaction. The largest effect sizes were observed in verbal memory in the placebo group (d = 0.69 in short-term, and d = 0.78 in long-term verbal memory). Increased IL-8 levels were found at 36-week follow-up in both Citrus peel extract and placebo groups (p for time = .010, d = 0.21, p time x treatment = .772). Adverse events were balanced between groups. CONCLUSIONS In this randomized clinical trial, long-term Citrus peel extract supplementation did not show cognitive benefits over placebo in participants with SCD, possibly due to high placebo response. These findings might have specific implications for designing future nutraceutical trials in individuals experiencing SCD. TRIAL REGISTRATION The trial has been registered at the United States National Library of Medicine at the National Institutes of Health Registry of Clinical Trials under the code NCT04744922 on February 9th, 2021 ( https://www. CLINICALTRIALS gov/ct2/show/NCT04744922 ).
Collapse
Affiliation(s)
- Samantha Galluzzi
- Laboratory Alzheimer's Neuroimaging & Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Moira Marizzoni
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy.
| | - Elena Gatti
- Laboratory Alzheimer's Neuroimaging & Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | | | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Francesco Epifano
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Giovanni B Frisoni
- Memory Center, Geneva University and University Hospitals, Geneva, Switzerland
| | - Salvatore Genovese
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Andrea Geviti
- Service of Statistics, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Lorenzo Marchetti
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Giovanni Sgrò
- Clinical Trial Service, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Claudio Singh Solorzano
- Laboratory Alzheimer's Neuroimaging & Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Michela Pievani
- Laboratory Alzheimer's Neuroimaging & Epidemiology, IRCCS Istituto Centro San Giovanni Di Dio Fatebenefratelli, Brescia, Italy
| | - Serena Fiorito
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
17
|
Lin YY, Chang WH, Hsieh SL, Cheng IHJ. The deficient CLEC5A ameliorates the behavioral and pathological deficits via the microglial Aβ clearance in Alzheimer's disease mouse model. J Neuroinflammation 2024; 21:273. [PMID: 39443966 PMCID: PMC11515658 DOI: 10.1186/s12974-024-03253-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: 05/15/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease that causes cognitive dysfunction in older adults. One of the AD pathological factors, β-Amyloid (Aβ), triggers inflammatory responses and phagocytosis of microglia. C-type lectin domain family 5 member A (CLEC5A) induces over-reactive inflammatory responses in several virus infections. Yet, the role of CLEC5A in AD progression remains unknown. This study aimed to elucidate the contribution of CLEC5A to Aβ-induced microglial activation and behavioral deficits. METHODS The AD mouse model was crossed with Clec5a knockout mice for subsequent behavioral and pathological tests. The memory deficit was revealed by the Morris water maze, while the nociception abnormalities were examined by the von Frey filament and hotplate test. The Aβ deposition and microglia recruitment were identified by ELISA and immunohistochemistry. The inflammatory signals were identified by ELISA and western blotting. In the Clec5a knockdown microglial cell model and Clec5a knockout primary microglia, the microglial phagocytosis was revealed using the fluorescent-labeled Aβ. RESULTS The AD mice with Clec5a knockout improved Aβ-induced memory deficit and abnormal nociception. These mice have reduced Aβ deposition and increased microglia coverage surrounding the amyloid plaque, suggesting the involvement of CLEC5A in AD progression and Aβ clearance. Moreover, the phagocytosis was also increased in the Aβ-stressed Clec5a knockdown microglial cell lines and Clec5a knockout primary microglia. CONCLUSION The Clec5a knockout ameliorates AD-like deficits by modulating microglial Aβ clearance. This study implies that targeting microglial Clec5a could offer a promising approach to mitigate AD progression.
Collapse
MESH Headings
- Animals
- Lectins, C-Type/metabolism
- Lectins, C-Type/deficiency
- Lectins, C-Type/genetics
- Microglia/metabolism
- Microglia/pathology
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Alzheimer Disease/genetics
- Mice
- Amyloid beta-Peptides/metabolism
- Disease Models, Animal
- Mice, Knockout
- Mice, Inbred C57BL
- Male
- Mice, Transgenic
- Maze Learning/physiology
- Phagocytosis
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
Collapse
Affiliation(s)
- Yu-Yi Lin
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wen-Han Chang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan.
- Institute of Clinical Medicine, Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Irene Han-Juo Cheng
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
18
|
Hu Y, Cui J, Sun J, Liu X, Gao S, Mei X, Wu C, Tian H. A novel biomimetic nanovesicle containing caffeic acid-coupled carbon quantum dots for the the treatment of Alzheimer's disease via nasal administration. J Nanobiotechnology 2024; 22:642. [PMID: 39425199 PMCID: PMC11490022 DOI: 10.1186/s12951-024-02912-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by progressive cognitive and physical impairment. Neuroinflammation is related to AD, and the misfolding and aggregation of amyloid protein in the brain creates an inflammatory microenvironment. Microglia are the predominant contributors to neuroinflammation, and abnormal activation of microglia induces the release of a large amount of inflammatory factors, promotes neuronal apoptosis, and leads to cognitive impairment. In this study, we used microglial membranes containing caffeic acid-coupled carbon quantum dots to prepare a novel biomimetic nanocapsule (CDs-CA-MGs) for the treatment of AD. The application of CDs-CA-MGs via nasal administration can bypass the blood‒brain barrier (BBB) and directly target the site of inflammation. After treatment with CDs-CA-MGs, AD mice showed reduced inflammation in the brain, decreased neuronal apoptosis, and significantly improved learning and memory abilities. In addition, CDs-CA-MGs affect inflammation-related JAK-STAT and Toll-like receptor signaling pathways in AD mice. CDs-CA-MGs significantly downregulated interleukins (IL-1β and IL-6) and tumor necrosis factor (TNF-α). This finding suggested that CDs-CA-MGs may improve cognitive impairment by modulating inflammatory responses. In conclusion, the use of CDs-CA-MGs provides a possible therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Yu Hu
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Jingwen Cui
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Junpeng Sun
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Xiaobang Liu
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Shuang Gao
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Xifan Mei
- Liaoning Vocational College of Medicine, Shenyang, Liaoning, 110101, China.
| | - Chao Wu
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- Pharmacy School, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| | - He Tian
- School of Basic Medicine, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
- Liaoning Provincial Collaborative Innovation Center for Medical Testing and Drug Research, Jinzhou Medical University, Jinzhou, Liaoning, 121001, China.
| |
Collapse
|
19
|
Childs R, Karamacoska D, Lim CK, Steiner-Lim GZ. "Let's talk about sex, inflammaging, and cognition, baby": A meta-analysis and meta-regression of 106 case-control studies on mild cognitive impairment and Alzheimer's disease. Brain Behav Immun Health 2024; 40:100819. [PMID: 39161876 PMCID: PMC11331696 DOI: 10.1016/j.bbih.2024.100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/21/2024] [Accepted: 07/06/2024] [Indexed: 08/21/2024] Open
Abstract
Background Chronic inflammation is recognised as an important component of Alzheimer's disease (AD), yet its relationship with cognitive decline, sex-differences, and age is not well understood. This study investigated the relationship between inflammatory markers, cognition, sex, and age in individuals with mild cognitive impairment (MCI) and AD. Methods A systematic review was performed to identify case-control studies which measured cognitive function and inflammatory markers in serum, plasma, and cerebrospinal fluid in individuals with MCI or AD compared with healthy control (HC) participants. Meta-analysis was performed with Hedges' g calculated in a random effects model. Meta-regression was conducted using age, sex, and mini-mental status exam (MMSE) values. Results A total of 106 studies without a high risk of bias were included in the meta-analysis including 18,145 individuals: 5625 AD participants, 3907 MCI participants, and 8613 HC participants. Combined serum and plasma meta-analysis found that IL1β, IL6, IL8, IL18, CRP, and hsCRP were significantly raised in individuals with AD compared to HC. In CSF, YKL40, and MCP-1 were raised in AD compared to HC. YKL40 was also raised in MCI compared to HC. Meta-regression analysis highlighted several novel findings: MMSE was negatively correlated with IL6 and positively correlated with IL1α in AD, while in MCI studies, MMSE was negatively correlated with IL8 and TNFα. Meta-regression also revealed sex-specific differences in levels of IL1α, IL4, IL6, IL18, hsCRP, MCP-1, and YKL-40 across AD and MCI studies, and age was found to account for heterogeneity of CRP, MCP-1, and IL4 in MCI and AD. Conclusion Elevated levels of IL6 and YKL40 may reflect microglial inflammatory activity in both MCI and AD. Systemic inflammation may interact with the central nervous system, as poor cognitive function in individuals with AD and MCI was associated with higher levels of serum and plasma proinflammatory cytokines IL6 and TNFα. Moreover, variations of systemic inflammation between males and females may be modulated by sex-specific hormonal changes, such as declining oestrogen levels in females throughout the menopause transition. Longitudinal studies sampling a range of biospecimen types are needed to elucidate the nuances of the relationship between inflammation and cognition in individuals with MCI and AD, and understand how systemic and central inflammation differentially impact cognitive function.
Collapse
Affiliation(s)
- Ryan Childs
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Diana Karamacoska
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Chai K. Lim
- Faculty of Medicine, Health, and Human Sciences, Macquarie University, Macquarie Park NSW, 2190, Australia
| | | |
Collapse
|
20
|
Seong SJ, Kim KW, Song JY, Park KJ, Jo YT, Han JH, Yoo KH, Jo HJ, Hwang JY. Inflammatory Cytokines and Cognition in Alzheimer's Disease and Its Prodrome. Psychiatry Investig 2024; 21:1054-1064. [PMID: 39465234 PMCID: PMC11513865 DOI: 10.30773/pi.2024.0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/03/2024] [Accepted: 07/05/2024] [Indexed: 10/29/2024] Open
Abstract
OBJECTIVE The aim of this study was to investigate the association between blood levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) and cognitive impairments among elderly individuals. METHODS Peripheral concentration of TNF-α and IL-6 were measured in all subjects. To assess individual cognitive function, the Consortium to Establish a Registry for Alzheimer's Disease Neuropsychological Assessment Battery (CERAD-NP) was used, and standardized scores (z-scores) were calculated for each test. Cytokine levels were compared between the diagnostic groups, and correlations between blood inflammatory factor levels and z-scores were analyzed. RESULTS The 37 participants included 8 patients with Alzheimer's disease (AD), 15 subjects with mild cognitive impairment (MCI), and 14 cognitively healthy controls. TNF-α and IL-6 levels were higher in patients with AD than in healthy controls. TNF-α levels were higher in the AD group than in the MCI group. However, after adjusting for age, the associations between diagnosis and TNF-α and IL-6 were not significant. The higher the plasma IL-6 level, the lower the z-scores on the Boston Naming Test, Word List Learning, Word List Recognition, and Constructional Recall. The higher the serum TNF-α level, the lower the z-scores on the Word List Learning and Constructional Recall. Negative correlation between serum TNF-α level and the z-score on Word List Learning remained significant when age was adjusted. CONCLUSION The difference in the blood levels of TNF-α and IL-6 between the diagnostic groups may be associated with aging. However, elevated TNF-α levels were associated with worse immediate memory performance, even after adjusting for age.
Collapse
Affiliation(s)
- Su Jeong Seong
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Ki Woong Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Brain and Cognitive Science, Seoul National University, College of Natural Sciences, Seoul, Republic of Korea
- Department of Neuropsychiatry, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Joo Yun Song
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Kee Jeong Park
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Young Tak Jo
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Jae Hyun Han
- Department of Psychiatry, Soonchunhyang University Cheonan Hospital, College of Medicine, Cheonan, Republic of Korea
| | - Ka Hee Yoo
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Hyun Jun Jo
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Jae Yeon Hwang
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| |
Collapse
|
21
|
Joshi A, Giorgi FM, Sanna PP. Transcriptional Patterns in Stages of Alzheimer's Disease Are Cell-Type-Specific and Partially Converge with the Effects of Alcohol Use Disorder in Humans. eNeuro 2024; 11:ENEURO.0118-24.2024. [PMID: 39299805 PMCID: PMC11485264 DOI: 10.1523/eneuro.0118-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
Advances in single-cell technologies have led to the discovery and characterization of new brain cell types, which in turn lead to a better understanding of the pathogenesis of Alzheimer's disease (AD). Here, we present a detailed analysis of single-nucleus (sn)RNA-seq data for three stages of AD from middle temporal gyrus and compare it with snRNA-seq data from the prefrontal cortices from individuals with alcohol use disorder (AUD). We observed a significant decrease in both inhibitory and excitatory neurons, in general agreement with previous reports. We observed several cell-type-specific gene expressions and pathway dysregulations that delineate AD stages. Endothelial and vascular leptomeningeal cells showed the greatest degree of gene expression changes. Cell-type-specific evidence of neurodegeneration was seen in multiple neuronal cell types particularly in somatostatin and Layer 5 extratelencephalic neurons, among others. Evidence of inflammatory responses was seen in non-neuronal cells, particularly in intermediate and advanced AD. We observed common perturbations in AD and AUD, particularly in pathways, like transcription, translation, apoptosis, autophagy, calcium signaling, neuroinflammation, and phosphorylation, that imply shared transcriptional pathogenic mechanisms and support the role of excessive alcohol intake in AD progression. Major AUD gene markers form and perturb a network of genes significantly associated with intermediate and advanced AD. Master regulator analysis from AUD gene markers revealed significant correlation with advanced AD of transcription factors that have implications in intellectual disability, neuroinflammation, and other neurodegenerative conditions, further suggesting a shared nexus of transcriptional changes between AD and AUD.
Collapse
Affiliation(s)
- Arpita Joshi
- The Scripps Research Institute, San Diego, California 92117
| | - Federico Manuel Giorgi
- The Scripps Research Institute, San Diego, California 92117
- University of Bologna, Bologna 40136, Italy
| | | |
Collapse
|
22
|
Yao L, Li MY, Wang KC, Liu YZ, Zheng HZ, Zhong Z, Ma SQ, Yang HM, Sun MM, He M, Huang HP, Wang HF. Abnormal resting-state functional connectivity of hippocampal subregions in type 2 diabetes mellitus-associated cognitive decline. Front Psychiatry 2024; 15:1360623. [PMID: 39376966 PMCID: PMC11456530 DOI: 10.3389/fpsyt.2024.1360623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
Objective Type 2 diabetes mellitus (T2DM) over time predisposes to inflammatory responses and abnormalities in functional brain networks that damage learning, memory, or executive function. The hippocampus is a key region often reporting connectivity abnormalities in memory disorders. Here, we investigated peripheral inflammatory responses and resting-state functional connectivity (RSFC) changes characterized of hippocampal subregions in type 2 diabetes-associated cognitive decline (T2DACD). Methods The study included 16 patients with T2DM, 16 patients with T2DACD and 25 healthy controls (HCs). Subjects were assessed for cognitive performance, tested for the expression of inflammatory factors IL-6, IL-10 and TNF-α in peripheral serum, underwent resting-state functional magnetic resonance imaging scans, and analyzed for RSFC using the hippocampal subregions as seeds. We also calculated the correlation between cognitive performance and RSFC of hippocampal subregion, and analyzed the significantly altered RSFC values of T2DACD for Receiver Operating Characteristic (ROC) analysis. Results T2DACD patients showed a decline in their ability to complete cognitive assessment scales and experimental paradigms, and T2DM did not show abnormal cognitive performance. IL-6 expression was increased in peripheral serum in both T2DACD and T2DM. Compared with HCs, T2DACD showed abnormalities RSFC of the left anterior hippocampus with left precentral gyrus and left angular gyrus. T2DM showed abnormalities RSFC of the left middle hippocampus with right medial frontal gyrus, right anterior and middle hippocampus with left precuneus, left anterior hippocampus with right precuneus and right posterior middle temporal gyrus. Compared with T2DM, T2DACD showed abnormalities RSFC of the left posterior hippocampus and right middle hippocampus with left precuneus. In addition, RSFC in the left posterior hippocampus with left precuneus of T2DACD was positively correlated with Flanker conflict response time (r=0.766, P=0.001). In the ROC analysis, the significantly altered RSFC values of T2DACD achieved significant performance. Conclusions T2DACD showed a significant decrease in attentional inhibition and working memory, peripheral pro-inflammatory response increased, and abnormalities RSFC of the hippocampal subregions with default mode network and sensory-motor network. T2DM did not show a significant cognitive decline, but peripheral pro-inflammatory response increased and abnormalities RSFC of the hippocampus subregions occurred in the brain. In addition, the left precuneus may be a key brain region in the conversion of T2DM to T2DACD. The results of this study may provide a basis for the preliminary diagnosis of T2DACD.
Collapse
Affiliation(s)
- Lin Yao
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meng-Yuan Li
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Kang-Cheng Wang
- College of Psychology, Shandong Normal University, Jinan, Shandong, China
| | - Yan-Ze Liu
- Acupuncture and Tuina Center, The Third Affiliated Clinical Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hai-Zhu Zheng
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhen Zhong
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Shi-Qi Ma
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hong-Mei Yang
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meng-Meng Sun
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Min He
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hai-Peng Huang
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hong-Feng Wang
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
23
|
Shao F, Hu J, Zhang P, Akarapipad P, Park JS, Lei H, Hsieh K, Wang TH. Enhanced CRISPR/Cas-Based Immunoassay through Magnetic Proximity Extension and Detection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.06.24313206. [PMID: 39314939 PMCID: PMC11419220 DOI: 10.1101/2024.09.06.24313206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas-associated systems have recently emerged as a focal point for developing next-generation molecular diagnosis, particularly for nucleic acid detection. However, the detection of proteins is equally critical across diverse applications in biology, medicine, and the food industry, especially for diagnosing and prognosing diseases like cancer, Alzheimer's and cardiovascular conditions. Despite recent efforts to adapt CRISPR/Cas systems for protein detection with immunoassays, these methods typically achieved sensitivity only in the femtomolar to picomolar range, underscoring the need for enhanced detection capabilities. To address this, we developed CRISPR-AMPED, an innovative CRISPR/Cas-based immunoassay enhanced by magnetic proximity extension and detection. This approach combines proximity extension assay (PEA) with magnetic beads that converts protein into DNA barcodes for quantification with effective washing steps to minimize non-specific binding and hybridization, therefore reducing background noise and increasing detection sensitivity. The resulting DNA barcodes are then detected through isothermal nucleic acid amplification testing (NAAT) using recombinase polymerase amplification (RPA) coupled with the CRISPR/Cas12a system, replacing the traditional PCR. This integration eliminates the need for thermocycling and bulky equipment, reduces amplification time, and provides simultaneous target and signal amplification, thereby significantly boosting detection sensitivity. CRISPR-AMPED achieves attomolar level sensitivity, surpassing ELISA by over three orders of magnitude and outperforming existing CRISPR/Cas-based detection systems. Additionally, our smartphone-based detection device demonstrates potential for point-of-care applications, and the digital format extends dynamic range and enhances quantitation precision. We believe CRISPR-AMPED represents a significant advancement in the field of protein detection.
Collapse
|
24
|
Daniilidou M, Holleman J, Hagman G, Kåreholt I, Aspö M, Brinkmalm A, Zetterberg H, Blennow K, Solomon A, Kivipelto M, Sindi S, Matton A. Neuroinflammation, cerebrovascular dysfunction and diurnal cortisol biomarkers in a memory clinic cohort: Findings from the Co-STAR study. Transl Psychiatry 2024; 14:364. [PMID: 39251589 PMCID: PMC11385239 DOI: 10.1038/s41398-024-03072-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024] Open
Abstract
Cortisol dysregulation, neuroinflammation, and cerebrovascular dysfunction are biological processes that have been separately shown to be affected in Alzheimer's disease (AD). Here, we aimed to identify biomarker signatures reflecting these pathways in 108 memory clinic patients with subjective cognitive decline (SCD, N = 40), mild cognitive impairment (MCI, N = 39), and AD (N = 29). Participants were from the well-characterized Cortisol and Stress in Alzheimer's Disease (Co-STAR) cohort, recruited at Karolinska University Hospital. Salivary diurnal cortisol measures and 41 CSF proteins were analyzed. Principal component analysis was applied to identify combined biosignatures related to AD pathology, synaptic loss, and neuropsychological assessments, in linear regressions adjusted for confounders, such as age, sex, education and diagnosis. We found increased CSF levels of C-reactive protein (CRP), interferon γ-inducible protein (IP-10), thymus and activation-regulated chemokine (TARC), intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) in MCI patients. Further, markers of cortisol dysregulation (flattened salivary cortisol awakening response and flattened cortisol slope) correlated with increased levels of placental growth factor (PlGF), IP-10, and chitinase 3-like 1 (YKL-40) in the total cohort. A biosignature composed of cortisol awakening response, cortisol slope, and CSF IL-6 was downregulated in AD patients. Moreover, biomarker signatures reflecting overlapping pathophysiological processes of neuroinflammation and vascular injury were associated with AD pathology, synaptic loss, and worsened processing speed. Our findings suggest an early dysregulation of immune and cerebrovascular processes during the MCI stage and provide insights into the interrelationship of chronic stress and neuroinflammation in AD.
Collapse
Affiliation(s)
- Makrina Daniilidou
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Jasper Holleman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Göran Hagman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Ingemar Kåreholt
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Institute of Gerontology, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Malin Aspö
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Alina Solomon
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Shireen Sindi
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Anna Matton
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
25
|
Luo Y, Zhu J, Hu Z, Luo W, Du X, Hu H, Peng S. Progress in the Pathogenesis of Diabetic Encephalopathy: The Key Role of Neuroinflammation. Diabetes Metab Res Rev 2024; 40:e3841. [PMID: 39295168 DOI: 10.1002/dmrr.3841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/29/2024] [Accepted: 06/27/2024] [Indexed: 09/21/2024]
Abstract
Diabetic encephalopathy (DE) is a severe complication that occurs in the central nervous system (CNS) and leads to cognitive impairment. DE involves various pathophysiological processes, and its pathogenesis is still unclear. This review summarised current research on the pathogenesis of diabetic encephalopathy, which involves neuroinflammation, oxidative stress, iron homoeostasis, blood-brain barrier disruption, altered gut microbiota, insulin resistance, etc. Among these pathological mechanisms, neuroinflammation has been focused on. This paper summarises some of the molecular mechanisms involved in neuroinflammation, including the Mammalian Target of Rapamycin (mTOR), Lipocalin-2 (LCN-2), Pyroptosis, Advanced Glycosylation End Products (AGEs), and some common pro-inflammatory factors. In addition, we discuss recent advances in the study of potential therapeutic targets for the treatment of DE against neuroinflammation. The current research on the pathogenesis of DE is progressing slowly, and more research is needed in the future. Further study of neuroinflammation as a mechanism is conducive to the discovery of more effective treatments for DE in the future.
Collapse
Affiliation(s)
- Yifan Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jinxi Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Clinical Medicine, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaohong Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haijun Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
26
|
Femiano C, Bruno A, Gilio L, Buttari F, Dolcetti E, Galifi G, Azzolini F, Borrelli A, Furlan R, Finardi A, Musella A, Mandolesi G, Storto M, Centonze D, Stampanoni Bassi M. Inflammatory signature in amyotrophic lateral sclerosis predicting disease progression. Sci Rep 2024; 14:19796. [PMID: 39187524 PMCID: PMC11347586 DOI: 10.1038/s41598-024-67165-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/09/2024] [Indexed: 08/28/2024] Open
Abstract
Experimental studies identified a role of neuroinflammation in the pathogenesis of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). However, the role of inflammatory molecules as diagnostic and prognostic biomarkers in patients with ALS is unclear. In this cross-sectional study, the cerebrospinal fluid (CSF) levels of a set of inflammatory cytokines and chemokines were analyzed in 56 newly diagnosed ALS patients and in 47 age- and sex-matched control patients without inflammatory or degenerative neurological disorders. The molecules analyzed included: interleukin (IL)-1β, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-17, granulocyte colony stimulating factor (GCSF), macrophage inflammatory protein (MIP)-1a, MIP-1b, tumor necrosis factors (TNF), eotaxin. Principal component analysis (PCA) was used to explore possible associations between CSF molecules and ALS diagnosis. In addition, we analyzed the association between CSF cytokine profiles and clinical characteristics, including the disease progression rate score, and peripheral inflammation assessed using the Neutrophil-to-lymphocyte ratio (NLR). PCA identified six principal components (PCs) explaining 70.67% of the total variance in the CSF cytokine set. The principal component (PC1) explained 26.8% of variance and showed a positive load with CSF levels of IL-9, IL-4, GCSF, IL-7, IL-17, IL-13, IL-6, IL-1β, TNF, and IL-2. Logistic regression showed a significant association between PC1 and ALS diagnosis. In addition, in ALS patients, the same component was significantly associated with higher disease progression rate score and positively correlated with NLR. CSF inflammatory activation in present in ALS at the time of diagnosis and may characterize patients at higher risk for disease progression.
Collapse
Affiliation(s)
| | - Antonio Bruno
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Luana Gilio
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
- Faculty of Psychology, International Telematic University UNINETTUNO, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | | - Giovanni Galifi
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | | | | | - Roberto Furlan
- Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Roma San Raffaele, Rome, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Roma San Raffaele, Rome, Italy
| | | | - Diego Centonze
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy.
- Department of Systems Medicine, Tor Vergata University, Rome, Italy.
| | | |
Collapse
|
27
|
Chaves JCS, Milton LA, Stewart R, Senapati T, Rantanen LM, Wasielewska JM, Lee S, Hernández D, McInnes L, Quek H, Pébay A, Donnelly PS, White AR, Oikari LE. Differential Cytokine Responses of APOE3 and APOE4 Blood-brain Barrier Cell Types to SARS-CoV-2 Spike Proteins. J Neuroimmune Pharmacol 2024; 19:22. [PMID: 38771543 DOI: 10.1007/s11481-024-10127-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 05/13/2024] [Indexed: 05/22/2024]
Abstract
SARS-CoV-2 spike proteins have been shown to cross the blood-brain barrier (BBB) in mice and affect the integrity of human BBB cell models. However, the effects of SARS-CoV-2 spike proteins in relation to sporadic, late onset, Alzheimer's disease (AD) risk have not been extensively investigated. Here we characterized the individual and combined effects of SARS-CoV-2 spike protein subunits S1 RBD, S1 and S2 on BBB cell types (induced brain endothelial-like cells (iBECs) and astrocytes (iAstrocytes)) generated from induced pluripotent stem cells (iPSCs) harboring low (APOE3 carrier) or high (APOE4 carrier) relative Alzheimer's risk. We found that treatment with spike proteins did not alter iBEC integrity, although they induced the expression of several inflammatory cytokines. iAstrocytes exhibited a robust inflammatory response to SARS-CoV-2 spike protein treatment, with differences found in the levels of cytokine secretion between spike protein-treated APOE3 and APOE4 iAstrocytes. Finally, we tested the effects of potentially anti-inflammatory drugs during SARS-CoV-2 spike protein exposure in iAstrocytes, and discovered different responses between spike protein treated APOE4 iAstrocytes and APOE3 iAstrocytes, specifically in relation to IL-6, IL-8 and CCL2 secretion. Overall, our results indicate that APOE3 and APOE4 iAstrocytes respond differently to anti-inflammatory drug treatment during SARS-CoV-2 spike protein exposure with potential implications to therapeutic responses.
Collapse
Affiliation(s)
- Juliana C S Chaves
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane (QLD), Australia
- Queensland University of Technology, Brisbane (QLD), Australia
| | - Laura A Milton
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane (QLD), Australia
| | - Romal Stewart
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane (QLD), Australia
| | | | - Laura M Rantanen
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane (QLD), Australia
- Queensland University of Technology, Brisbane (QLD), Australia
| | - Joanna M Wasielewska
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane (QLD), Australia
- Faculty of Medicine, The University of Queensland, Brisbane (QLD), Australia
| | - Serine Lee
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane (QLD), Australia
| | - Damián Hernández
- Department of Anatomy and Physiology, The University of Melbourne, Parkville (VIC), Australia
| | - Lachlan McInnes
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville (VIC), Australia
| | - Hazel Quek
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane (QLD), Australia
- Queensland University of Technology, Brisbane (QLD), Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane (QLD), Australia
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville (VIC), Australia
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville (VIC), Australia
| | - Paul S Donnelly
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville (VIC), Australia
| | - Anthony R White
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane (QLD), Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane (QLD), Australia
| | - Lotta E Oikari
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane (QLD), Australia.
- Queensland University of Technology, Brisbane (QLD), Australia.
| |
Collapse
|
28
|
Holleman J, Daniilidou M, Kåreholt I, Aspö M, Hagman G, Udeh-Momoh CT, Spulber G, Kivipelto M, Solomon A, Matton A, Sindi S. Diurnal cortisol, neuroinflammation, and neuroimaging visual rating scales in memory clinic patients. Brain Behav Immun 2024; 118:499-509. [PMID: 38503394 DOI: 10.1016/j.bbi.2024.03.024] [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: 10/30/2023] [Revised: 02/18/2024] [Accepted: 03/16/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Neuroinflammation is a hallmark of the Alzheimer's disease (AD) pathogenic process. Cortisol dysregulation may increase AD risk and is related to brain atrophy. This cross-sectional study aims to examine interactions of cortisol patterns and neuroinflammation markers in their association with neuroimaging correlates. METHOD 134 participants were recruited from the Karolinska University Hospital memory clinic (Stockholm, Sweden). Four visual rating scales were applied to magnetic resonance imaging or computed tomography scans: medial temporal lobe atrophy (MTA), global cortical atrophy (GCA), white matter lesions (WML), and posterior atrophy. Participants provided saliva samples for assessment of diurnal cortisol patterns, and underwent lumbar punctures for cerebrospinal fluid (CSF) sampling. Three cortisol measures were used: the cortisol awakening response, total daily output, and the ratio of awakening to bedtime levels. Nineteen CSF neuroinflammation markers were categorized into five composite scores: proinflammatory cytokines, other cytokines, angiogenesis markers, vascular injury markers, and glial activation markers. Ordinal logistic regressions were conducted to assess associations between cortisol patterns, neuroinflammation scores, and visual rating scales, and interactions between cortisol patterns and neuroinflammation scores in relation to visual rating scales. RESULT Higher levels of angiogenesis markers were associated with more severe WML. Some evidence was found for interactions between dysregulated diurnal cortisol patterns and greater neuroinflammation-related biomarkers in relation to more severe GCA and WML. No associations were found between cortisol patterns and visual rating scales. CONCLUSION This study suggests an interplay between diurnal cortisol patterns and neuroinflammation in relation to brain structure. While this cross-sectional study does not provide information on causality or temporality, these findings suggest that neuroinflammation may be involved in the relationship between HPA-axis functioning and AD.
Collapse
Affiliation(s)
- Jasper Holleman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden.
| | - Makrina Daniilidou
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ingemar Kåreholt
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden; Institute of Gerontology, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Malin Aspö
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden
| | - Göran Hagman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden
| | - Chinedu T Udeh-Momoh
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK; Division of Public Health Sciences, Wake Forest University School of Medicine, North Carolina, USA; Brain and Mind Institute, Aga Khan University, Nairobi, Kenya
| | - Gabriela Spulber
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Alina Solomon
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Theme Inflammation and Aging. Karolinska University Hospital, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK; Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Anna Matton
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Division of Neurogeriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK
| | - Shireen Sindi
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Karolinska Vägen 37A - QA32, 171 64 Solna, Stockholm, Sweden; Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, UK
| |
Collapse
|
29
|
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) is an all-encompassing term that describes cognitive impairment due to cerebrovascular origins. With the advancement of imaging and pathological studies, we now understand that VCID is often comorbid with Alzheimer disease. While researchers in the Alzheimer disease field have been working for years to establish and test blood-based biomarkers for Alzheimer disease diagnosis, prognosis, clinical therapy discovery, and early detection, blood-based biomarkers for VCID are in their infancy and also face challenges. VCID is heterogeneous, comprising many different pathological entities (ischemic, or hemorrhagic), and spatial and temporal differences (acute or chronic). This review highlights pathways that are aiding the search for sensitive and specific blood-based cerebrovascular dysfunction markers, describes promising candidates, and explains ongoing initiatives to discover blood-based VCID biomarkers.
Collapse
Affiliation(s)
- Kate E. Foley
- Stark Neurosciences Research Institute, Indiana University, Indianapolis IN, USA
- Department of Neurology, School of Medicine, Indiana University, Indianapolis IN, USA
| | - Donna M. Wilcock
- Stark Neurosciences Research Institute, Indiana University, Indianapolis IN, USA
- Department of Neurology, School of Medicine, Indiana University, Indianapolis IN, USA
| |
Collapse
|
30
|
Capogna E, Sørensen Ø, Watne LO, Roe J, Strømstad M, Idland AV, Halaas NB, Blennow K, Zetterberg H, Walhovd KB, Fjell AM, Vidal-Piñeiro D. Subtypes of brain change in aging and their associations with cognition and Alzheimer's disease biomarkers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583291. [PMID: 38496633 PMCID: PMC10942348 DOI: 10.1101/2024.03.04.583291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Structural brain changes underly cognitive changes in older age and contribute to inter-individual variability in cognition. Here, we assessed how changes in cortical thickness, surface area, and subcortical volume, are related to cognitive change in cognitively unimpaired older adults using structural magnetic resonance imaging (MRI) data-driven clustering. Specifically, we tested (1) which brain structural changes over time predict cognitive change in older age (2) whether these are associated with core cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers phosphorylated tau (p-tau) and amyloid-β (Aβ42), and (3) the degree of overlap between clusters derived from different structural features. In total 1899 cognitively healthy older adults (50 - 93 years) were followed up to 16 years with neuropsychological and structural MRI assessments, a subsample of which (n = 612) had CSF p-tau and Aβ42 measurements. We applied Monte-Carlo Reference-based Consensus clustering to identify subgroups of older adults based on structural brain change patterns over time. Four clusters for each brain feature were identified, representing the degree of longitudinal brain decline. Each brain feature provided a unique contribution to brain aging as clusters were largely independent across modalities. Cognitive change and baseline cognition were best predicted by cortical area change, whereas higher levels of p-tau and Aβ42 were associated with changes in subcortical volume. These results provide insights into the link between changes in brain morphology and cognition, which may translate to a better understanding of different aging trajectories.
Collapse
Affiliation(s)
- Elettra Capogna
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Oslo, Norway
| | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Oslo, Norway
| | - Leiv Otto Watne
- Department of Geriatric Medicine, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - James Roe
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Oslo, Norway
| | - Marie Strømstad
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Oslo, Norway
| | - Ane Victoria Idland
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Nathalie Bodd Halaas
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Campus UllevÅl, University of Oslo, Oslo, Norway
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kristine Beate Walhovd
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Oslo, Norway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Anders Martin Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Oslo, Norway
- Computational Radiology and Artificial Intelligence, Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Didac Vidal-Piñeiro
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Oslo, Norway
| |
Collapse
|
31
|
Yao Y, Zhu S, Ni J, Wei M, Li T, Long S, Shi J, Tian J. Gout or Hyperuricemia and Dementia Risk: A Meta-Analysis of Observational Studies. J Alzheimers Dis 2024; 101:417-427. [PMID: 39240635 DOI: 10.3233/jad-240076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Background As a natural antioxidant, uric acid has neuroprotective effects. The association between uric acid levels and dementia risk was reported by previous studies. However, recently published studies showed that the relationship between uric acid and dementia risk might be heterogeneous in dementia subtypes. Objective This study aimed to clarify the relationship between hyperuricemia (or gout) and dementia. Methods The PubMed and Web of Science databases were systematically searched up to April 2024 to identify relevant studies. A meta-analysis was conducted using hazard ratios (HR) or odds ratios (OR) and 95% confidence interval (CI) as pooled indicators. Heterogeneity between the studies was examined using Cochran's Q statistic and I2 statistic. Subgroup analyses were conducted for gender and age. Stratification analysis, sensitivity analyses and meta-regression were conducted to explore possible explanations for heterogeneity. Publication bias was assessed by funnel plot and Egger's test. Results A total of 11 studies met the inclusion criteria including 2,928,152 participants were abstracted. Hyperuricemia (or gout) did not reduce the overall risk of dementia (OR/HR = 0.92, 95% CI: 0.81-1.05) and vascular dementia (OR/HR = 0.74, 95% CI: 0.53-1.05), but may have a protective effect against Alzheimer's disease (OR/HR = 0.82, 95% CI: 0.70-0.96). Subgroup analysis showed that a lower risk of dementia was observed in men (OR/HR = 0.83, 95% CI: 0.77-0.90) and patients whose age under 65 (OR/HR = 0.83, 95% CI: 0.72-0.95). Conclusions Patients with gout or hyperuricemia have a low risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Yirou Yao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shun Zhu
- 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
| | - Siwei Long
- 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
|
32
|
Abramova O, Zorkina Y, Ushakova V, Gryadunov D, Ikonnikova A, Fedoseeva E, Emelyanova M, Ochneva A, Morozova I, Pavlov K, Syunyakov T, Andryushchenko A, Savilov V, Kurmishev M, Andreuyk D, Shport S, Gurina O, Chekhonin V, Kostyuk G, Morozova A. Alteration of Blood Immune Biomarkers in MCI Patients with Different APOE Genotypes after Cognitive Training: A 1 Year Follow-Up Cohort Study. Int J Mol Sci 2023; 24:13395. [PMID: 37686198 PMCID: PMC10488004 DOI: 10.3390/ijms241713395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Many studies aim to detect the early phase of dementia. One of the major ways to achieve this is to identify corresponding biomarkers, particularly immune blood biomarkers. The objective of this study was to identify such biomarkers in patients with mild cognitive impairment (MCI) in an experiment that included cognitive training. A group of patients with MCI diagnoses over the age of 65 participated in the study (n = 136). Measurements of cognitive functions (using the Mini-Mental State Examination scale and Montreal Cognitive Assessment) and determination of 27 serum biomarkers were performed twice: on the first visit and on the second visit, one year after the cognitive training. APOE genotypes were also determined. Concentrations of EGF (F = 17; p = 0.00007), Eotaxin (F = 7.17; p = 0.008), GRO (F = 13.42; p = 0.0004), IL-8 (F = 8.16; p = 0.005), MCP-1 (F = 13.46; p = 0.0001) and MDC (F = 5.93; p = 0.016) increased after the cognitive training in MCI patients. All these parameters except IL-8 demonstrated a weak correlation with other immune parameters and were poorly represented in the principal component analysis. Differences in concentrations of IP-10, FGF-2, TGFa and VEGF in patients with MCI were associated with APOE genotype. Therefore, the study identified several immune blood biomarkers that could potentially be associated with changes in cognitive function.
Collapse
Affiliation(s)
- Olga Abramova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Yana Zorkina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Valeriya Ushakova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
- Biological Faculty, M.V. Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Dmitry Gryadunov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anna Ikonnikova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Elena Fedoseeva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Marina Emelyanova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Aleksandra Ochneva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Irina Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
| | - Konstantin Pavlov
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Timur Syunyakov
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- International Centre for Education and Research in Neuropsychiatry (ICERN), Samara State Medical University, 443016 Samara, Russia
| | - Alisa Andryushchenko
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
| | - Victor Savilov
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
| | - Marat Kurmishev
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
| | - Denis Andreuyk
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Biological Faculty, M.V. Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Svetlana Shport
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Olga Gurina
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Georgy Kostyuk
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Psychiatry, Federal State Budgetary Educational Institution of Higher Education “Moscow State University of Food Production”, Volokolamskoye Highway 11, 125080 Moscow, Russia
| | - Anna Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (O.A.); (Y.Z.); (V.U.); (A.O.); (I.M.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky per. 23, 119034 Moscow, Russia
| |
Collapse
|
33
|
Xu J, Gou S, Huang X, Zhang J, Zhou X, Gong X, Xiong J, Chi H, Yang G. Uncovering the Impact of Aggrephagy in the Development of Alzheimer's Disease: Insights Into Diagnostic and Therapeutic Approaches from Machine Learning Analysis. Curr Alzheimer Res 2023; 20:618-635. [PMID: 38141185 DOI: 10.2174/0115672050280894231214063023] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) stands as a widespread neurodegenerative disorder marked by the gradual onset of memory impairment, predominantly impacting the elderly. With projections indicating a substantial surge in AD diagnoses, exceeding 13.8 million individuals by 2050, there arises an urgent imperative to discern novel biomarkers for AD. METHODS To accomplish these objectives, we explored immune cell infiltration and the expression patterns of immune cells and immune function-related genes of AD patients. Furthermore, we utilized the consensus clustering method combined with aggrephagy-related genes (ARGs) for typing AD patients and categorized AD specimens into distinct clusters (C1, C2). A total of 272 candidate genes were meticulously identified through a combination of differential analysis and Weighted Gene Co-Expression Network Analysis (WGCNA). Subsequently, we applied three machine learning algorithms-namely random forest (RF), support vector machine (SVM), and generalized linear model (GLM)-to pinpoint a pathogenic signature comprising five genes associated with AD. To validate the predictive accuracy of these identified genes in discerning AD progression, we constructed nomograms. RESULTS Our analyses uncovered that cluster C2 exhibits a higher immune expression than C1. Based on the ROC(0.956). We identified five characteristic genes (PFKFB4, PDK3, KIAA0319L, CEBPD, and PHC2T) associated with AD immune cells and function. The nomograms constructed on the basis of these five diagnostic genes demonstrated effectiveness. In the validation group, the ROC values were found to be 0.760 and 0.838, respectively. These results validate the robustness and reliability of the diagnostic model, affirming its potential for accurate identification of AD. CONCLUSION Our findings not only contribute to a deeper understanding of the molecular mechanisms underlying AD but also offer valuable insights for drug development and clinical analysis. The limitation of our study is the limited sample size, and although AD-related genes were identified and some of the mechanisms elucidated, further experiments are needed to elucidate the more in-depth mechanisms of these characterized genes in the disease.
Collapse
Affiliation(s)
- Jiayu Xu
- School of Science, Minzu University of China, Beijing, China
| | - Siqi Gou
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xueyuan Huang
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jieying Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xuancheng Zhou
- Department of Psychiatry, Southwest Medical University, Luzhou, China
| | - Xiangjin Gong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Jingwen Xiong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Hao Chi
- School of Clinical Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, USA
| |
Collapse
|