1
|
Modiri A, Abdolmaleki Z, Paryani MR. The effect of rosuvastatin coated by nano-chitosan on developing hippocampus: association with hippocampal neurogenesis and memory in an Alzheimer's induced model of rats. Anat Cell Biol 2025; 58:61-75. [PMID: 39914828 PMCID: PMC11933804 DOI: 10.5115/acb.24.250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/26/2024] [Accepted: 11/01/2024] [Indexed: 03/22/2025] Open
Abstract
Statins are long known to be beneficial for neurodegenerative conditions, including Alzheimer's disease (AD). Also, nanoparticle (NP) drugs can better affect the target tissue in various diseases. Therefore, the aim of this study was surveying the effect of rosuvastatin (RZV) coated by nano-chitosan in an Alzheimer's (Alz) induced model of rats. We examined learning, memory, and hippocampal amyloid plaques and evaluate expression levels of calbindin, doublecortin (DCX), NeuroD1, neuronal nuclei (NeuN), and neurofilament. Forty rats were randomly divided into five various groups. AD was induced by injecting bilaterally with 1 μl of amyloid beta (Aβ) into the hippocampus. After confirmation of AD, RZV, or NP, or RZV+NP were administered gavage orally daily in rats for 30 days. Induction of AD significantly raised Aβ plaques and dead cells compared to the control group. Results of Morris water maze in the test day indicated that Alz+NP+RZV group significantly reduced escape latency and travelled distance, also significantly increased spending time compared to the Alz group (P<0.05). RZV significantly decreased Aβ plaque percentage and the number of apoptotic cells compared to the Alz group (P<0.05). In addition, NeuN and neurofilament protein expression and calbindin, DCX, and NeuroD1 genes expression increased in Alz+RZV and Alz+RZV+NP compared to the Alz group. RZV coated by nano-chitosan has good potential for reducing Aβ plaques and dead cells, increasing brain NeuN and neurofilament proteins and calbindin, DCX, and NeuroD1 genes, and improving learning and memory in Alz rats.
Collapse
Affiliation(s)
- Armita Modiri
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Zohreh Abdolmaleki
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Mohammad Reza Paryani
- Department of Veterinary Basic Sciences, Karaj Branch, Islamic Azad University, Karaj, Iran
| |
Collapse
|
2
|
Ray NR, Bradley J, Yilmaz E, Kizil C, Kurup JT, Martin ER, Klein HU, Kunkle BW, Bennett DA, De Jager PL, Beecham GW, Cruchaga C, Reitz C. Local genetic covariance analysis with lipid traits identifies novel loci for early-onset Alzheimer's Disease. PLoS Genet 2025; 21:e1011631. [PMID: 40096060 PMCID: PMC11984970 DOI: 10.1371/journal.pgen.1011631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 04/10/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
The genetic component of early-onset Alzheimer disease (EOAD), accounting for ~10% of all Alzheimer's disease (AD) cases, is largely unexplained. Recent studies suggest that EOAD may be enriched for variants acting in the lipid pathway. The current study examines the shared genetic heritability between EOAD and the lipid pathway using genome-wide multi-trait genetic covariance analyses. Summary statistics were obtained from the GWAS meta-analyses of EOAD by the Alzheimer's Disease Genetics Consortium (n=19,668) and five blood lipid traits by the Global Lipids Genetics Consortium (n=1,320,016). The significant results were compared between the EOAD and lipids GWAS and genetic covariance analyses were performed via SUPERGNOVA. Genes in linkage disequilibrium (LD) with top EOAD hits in identified regions of covariance with lipid traits were scored and ranked for causality by combining evidence from gene-based analysis, AD-risk scores incorporating transcriptomic and proteomic evidence, eQTL data, eQTL colocalization analyses, DNA methylation data, and single-cell RNA sequencing analyses. Direct comparison of GWAS results showed 5 loci overlapping between EOAD and at least one lipid trait harboring APOE, TREM2, MS4A4E, LILRA5, and LRRC25. Local genetic covariance analyses identified 3 regions of covariance between EOAD and at least one lipid trait. Gene prioritization nominated 3 likely causative genes at these loci: ANKDD1B, CUZD1, and MS4A64.The current study identified genetic covariance between EOAD and lipids, providing further evidence of shared genetic architecture and mechanistic pathways between the two traits.
Collapse
Affiliation(s)
- Nicholas R. Ray
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Joseph Bradley
- NeuroGenomics and Informatics Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Elanur Yilmaz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Caghan Kizil
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Jiji T. Kurup
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Eden R. Martin
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Hans-Ulrich Klein
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Brian W. Kunkle
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - David A. Bennett
- RUSH Alzheimer’s Disease Center, RUSH University, Chicago, Illinois, United States of America
| | - Philip L. De Jager
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | | | - Gary W. Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Carlos Cruchaga
- NeuroGenomics and Informatics Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Christiane Reitz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| |
Collapse
|
3
|
Rajab HA, Al-Kuraishy HM, Shokr MM, Al-Gareeb AI, Al-Harchan NA, Alruwaili M, Papadakis M, Alexiou A, Batiha GES. Statins for vascular dementia: A hype or hope. Neuroscience 2025; 567:45-55. [PMID: 39746645 DOI: 10.1016/j.neuroscience.2024.12.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Vascular dementia (VaD) is a second most common type of dementia subsequent to Alzheimer disease (AD). VaD is characterized by cognitive impairment and memory loss that may progress due to the development of cerebral amyloid angiopathy (CAA) a hallmark of AD. CAA triggers the progression of ischemic and hemorrhagic strokes with the subsequent the development of VaD and mixed dementia. Early diagnosis of patients with appropriate use of anti-inflammatory can prevent CAA-related inflammation and VaD development. Currently, there are no effective drugs in the management of VaD. Of note, cholesterol-lowering agent statins which are commonly used in patients with vascular diseases and dyslipidemia may affect the progression of VaD. Many previous studies highlighted the potential therapeutic efficacy of statins in treating VaD. Though, the underlying mechanisms of statins in prevention and treatment of VaD are not fully clarified. Consequently, this review aims to discuss the mechanistic role of statins in the management of VaD, and how statins may adversely affect the cognitive function in VaD patients.
Collapse
Affiliation(s)
- Hussein A Rajab
- Endocrinology Consultant, Medical School, Najran University, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Mustafa M Shokr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, Jabir Ibn Hayyan Medical University, Kufa, Iraq
| | - Nasser A Al-Harchan
- Department of Clinical Pharmacology, College of Dentistry, Al-Rasheed University, Baghdad, Iraq
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal, 42283, Germany.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia; Department of Research & Development, Funogen, Athens, Greece
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
4
|
Savulescu-Fiedler I, Dorobantu-Lungu LR, Dragosloveanu S, Benea SN, Dragosloveanu CDM, Caruntu A, Scheau AE, Caruntu C, Scheau C. The Cross-Talk Between the Peripheral and Brain Cholesterol Metabolisms. Curr Issues Mol Biol 2025; 47:115. [PMID: 39996836 PMCID: PMC11853762 DOI: 10.3390/cimb47020115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Cholesterol is an essential element for the development and normal function of the central nervous system. While peripheral cholesterol is influenced by liver metabolism and diet, brain cholesterol metabolism takes place in an isolated system due to the impermeability of the blood-brain barrier (BBB). However, cross-talk occurs between the brain and periphery, specifically through metabolites such as oxysterols that play key roles in regulating cholesterol balance. Several neurodegenerative conditions such as Alzheimer's disease or Parkinson's disease are considered to be affected by the loss of this balance. Also, the treatment of hypercholesterolemia needs to consider these discrete interferences between brain and peripheral cholesterol and the possible implications of each therapeutic approach. This is particularly important because of 27-hydroxycholesterol and 24-hydroxycholesterol, which can cross the BBB and are involved in cholesterol metabolism. This paper examines the metabolic pathways of cholesterol metabolism in the brain and periphery and focuses on the complex cross-talk between these metabolisms. Also, we emphasize the regulatory role of the BBB and the need for an integrated approach to cholesterol management.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Luiza-Roxana Dorobantu-Lungu
- Department of Cardiology, Emergency Institute for Cardiovascular Diseases “C.C. Iliescu”, 022328 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Departament of Infectious Diseases, National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania
| | - Christiana Diana Maria Dragosloveanu
- Department of Ophthalmology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
5
|
Li C, Zhang M, Du Y, Liu S, Li D, Zhang S, Ji F, Zhang J, Jiao J. Compromised cell competition exhausts neural stem cells pool. Cell Prolif 2024; 57:e13710. [PMID: 39010274 PMCID: PMC11628731 DOI: 10.1111/cpr.13710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/05/2024] [Accepted: 06/22/2024] [Indexed: 07/17/2024] Open
Abstract
Blood vessels play a crucial role in maintaining the stem cell niche in both tumours and developing organs. Cell competition is critical for tumour progression. We hypothesise that blood vessels may act as a regulator of this process. As a pioneer, the secretions of blood vessels regulate the intensity of cell competition, which is essential for tumour invasion and developmental organ extension. Brd4 expresses highly in endothelial cells within various tumours and is positively correlated with numerous invasive genes, making it an ideal focal point for further research on the relationship between blood vessels and cell competition. Our results indicated that the absence of endothelial Brd4 led to a reduction in neural stem cell mortality and compromised cell competition. Endothelial Brd4 regulated cell competition was dependent on Testican2. Testican2 was capable of depositing Sparc and acted as a suppressor of Sparc. Compromised cell competition resulted in the depletion of neural stem cells and accelerated brain ageing. Testican2 could rescue the run-off of neural stem cells and accelerate the turnover rate of neurons. AD patients show compromised cell competition. Through the cloning of a point mutant of Brd4 identified in a subset of AD patients, it was demonstrated that the mutant lacked the ability to promote cell competition. This study suggests a novel approach for treating age-related diseases by enhancing the intensity of cell competition.
Collapse
Affiliation(s)
- Chenxiao Li
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical UniversityZhanjiangChina
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of ScienceBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
| | - Mengtian Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of ScienceBeijingChina
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
| | - Yushan Du
- College of Basic Medicine, Qingdao UniversityQingdaoChina
| | - Shuang Liu
- Jiaozuo Hospital of Traditional Chinese MedicineHenanChina
| | - Da Li
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of ScienceBeijingChina
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
| | - Shukui Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of ScienceBeijingChina
| | - Fen Ji
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of ScienceBeijingChina
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical UniversityZhanjiangChina
| | - Jianwei Jiao
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of ScienceBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
| |
Collapse
|
6
|
Bi X, Cao N, He J. Recent advances in nanoenzymes for Alzheimer's disease treatment. Colloids Surf B Biointerfaces 2024; 244:114139. [PMID: 39121571 DOI: 10.1016/j.colsurfb.2024.114139] [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: 06/04/2024] [Revised: 07/14/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Alzheimer's disease (AD) remains one of the most challenging neurodegenerative disorders to treat, with oxidative stress playing a significant role in its pathology. Recent advancements in nanoenzymes technology offer a promising approach to mitigate this oxidative damage. Nanoenzymes, with their unique enzyme-mimicking activities, effectively scavenge reactive oxygen species and reduce oxidative stress, thereby providing neuroprotective effects. This review delves into the underlying mechanisms of AD, focusing on oxidative stress and its impact on disease progression. We explore the latest developments in nanoenzymes applications for AD treatment, highlighting their multifunctional capabilities and potential for targeted delivery to amyloid-beta plaques. Despite the exciting prospects, the clinical translation of nanoenzymes faces several challenges, including difficulties in brain targeting, consistent quality production, and ensuring safety and biocompatibility. We discuss these limitations in detail, emphasizing the need for rigorous evaluation and standardized protocols. This paper aims to provide a comprehensive overview of the current state of nanoenzymes research in AD, shedding light on both the opportunities and obstacles in the path towards effective clinical applications.
Collapse
Affiliation(s)
- Xiaojun Bi
- General Hospital of Northern Theater Command, Liaoning 110016, China
| | - Ning Cao
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing 400000, China
| | - Jingteng He
- General Hospital of Northern Theater Command, Liaoning 110016, China.
| |
Collapse
|
7
|
Kazibwe R, Rikhi R, Mirzai S, Ashburn NP, Schaich CL, Shapiro M. Do Statins Affect Cognitive Health? A Narrative Review and Critical Analysis of the Evidence. Curr Atheroscler Rep 2024; 27:2. [PMID: 39520593 PMCID: PMC11550230 DOI: 10.1007/s11883-024-01255-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW Statins are the first-line treatment for hypercholesterolemia and play a key role in the prevention of cardiovascular disease (CVD). Current studies report mixed effects of statins on cognitive health, including harmful, neutral, and protective outcomes. However, these ongoing controversies about the potential cognitive adverse effects of statins may compromise their use in CVD prevention. Several factors may influence how statins affect cognition, including the unique cholesterol homeostasis in the brain, the limited permeability of the blood-brain barrier to lipoproteins, and the varying lipophilicity of different statins. This review examines the evidence linking statins to cognitive function and considers the effect of different dosages and treatment durations. RECENT FINDINGS Earlier studies suggested cognitive disturbances with statins, but recent evidence does not strongly support a link between statins and cognitive impairment. In fact, observational studies suggest potential neuroprotective benefits, though biases like selection bias, confounding and reverse causation limit definitive conclusions. Two large randomized controlled trials, STAREE and PREVENTABLE, are underway, and their results are expected to address some of these gaps in the literature. Due to insufficient evidence in the current literature, well-designed randomized controlled trials are needed for a better understanding of statins' effects on cognition. More data is needed regarding statin type, dose intensity, and treatment duration, which may affect cognitive outcomes. Future studies are also needed to examine how statins may affect cognition in specific high-risk groups, such as individuals with mild cognitive impairment, diabetes, cardiovascular disease, or chronic kidney disease.
Collapse
Affiliation(s)
- Richard Kazibwe
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Rishi Rikhi
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Saeid Mirzai
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Nicklaus P Ashburn
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Christopher L Schaich
- Department of Emergency Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Michael Shapiro
- Center for Preventive Cardiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
8
|
Luo H, Hartikainen S, Lin J, Zhou H, Tapiainen V, Tolppanen AM. Predicting Alzheimer's disease from cognitive footprints in mid and late life: How much can register data and machine learning help? Int J Med Inform 2024; 190:105540. [PMID: 38972231 DOI: 10.1016/j.ijmedinf.2024.105540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 06/12/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND Real-world data with decades-long medical records are increasingly available alongside the growing adoption of machine learning in healthcare research. We evaluated the performance of machine learning models in predicting the risk of Alzheimer's disease (AD) using data from the Finnish national registers. METHODS We conducted a case-control study using data from the Finnish MEDALZ (Medication use and Alzheimer's disease) study. Altogether 56,741 individuals with incident AD diagnosis (age ≥ 65 years at diagnosis and born after 1922) and their 1:1 age-, sex-, and region of residence-matched controls were included. The association of risk factors, evaluated at different age periods (45-54, 55-64, 65+), and AD were assessed with logistic regression. Predictive accuracies of logistic regressions were compared with seven machine learning models (L1-regularized logistic regression, Naive bayes, Decision tree, Random Forest, Multilayer perceptron, XGBoost, and LightGBM). FINDINGS 63.5 % of cases and controls were females and the mean age was 79.1 (SD = 5.1). The strongest associations with AD were observed for head injuries at age 55-64 (OR, 95 % CI 1.33, 1.19-1.48) and 65+ (1.31, 1.23-1.40), followed by antidepressant use (1.30, 1.22-1.38) at 55-64 and antipsychotic use (1.27, 1.19-1.35) at 65+. The predictive accuracies of all models were low, with the best performance (AUC 0.603) observed in Random Forest for predicting AD onset at age 65-69. INTERPRETATION Although significant associations were identified between many risk factors and AD, the low predictive accuracies suggest that specialised healthcare diagnosis data is not sufficient for predicting AD and linkage with other data sources is needed.
Collapse
Affiliation(s)
- Hao Luo
- Department of Social Work and Social Administration, The University of Hong Kong, Hong Kong, China; Sau Po Centre on Ageing, The University of Hong Kong, Hong Kong, China; Kuopio Research Center of Geriatric Care, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Sirpa Hartikainen
- Kuopio Research Center of Geriatric Care, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Julian Lin
- Kuopio Research Center of Geriatric Care, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Huiquan Zhou
- Department of Psychiatry, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Vesa Tapiainen
- Kuopio Research Center of Geriatric Care, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Anna-Maija Tolppanen
- Kuopio Research Center of Geriatric Care, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
9
|
Wang W, Li X. Cognitive function in dyslipidemia patients: exploring the impact of statins. Front Neurol 2024; 15:1436010. [PMID: 39350969 PMCID: PMC11439768 DOI: 10.3389/fneur.2024.1436010] [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: 05/21/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Background Evidence regarding the relationship between the use of statins and cognitive outcomes presents varying findings. This study aims to analyze the relationship between sustained statin use and cognitive performance in dyslipidemia patients. Methods This study presents findings from the Beijing Ageing Brain Rejuvenation Initiative (BABRI) study, in which a cohort of community-dwelling dyslipidemia patients (Entire sample, N = 1,062, aged 50-86) was recruited. Participants were divided into two groups based on their sustained use statins (Statins group, N = 677) or not use any lipid-lowering agents (Untreated group, N = 385). Furthermore, the entire sample was stratified by age into the middle-aged sample (N = 451) and the older people sample (N = 611), following a similar categorization based on statin application. ANCOVA was used to evaluate the relationship between sustained statin use and cognitive function. Results Overall, in the total sample, the statins group demonstrated better cognition in overall cognition, memory, visuospatial ability, attention, executive function, and language domains compared to the untreated group. Moreover, the statins group only showed better performance in attention among the middle-aged sample. In the older people sample, statins group exhibited superior cognitive performance across various cognitive domains compared to untreated group. Conclusion Among dyslipidemia patients in Beijing community, sustained statin users exhibited superior cognitive function across all domains compared to untreated individuals, with particularly noticeable improvements among those aged 65 and above. These findings underscore the protective effect of statins on cognitive function in dyslipidemia patients, highlighting significant benefits for the older people population.
Collapse
Affiliation(s)
- Wenxiao Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- School of System Science, Beijing Normal University, Beijing, China
| | - Xin Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- BABRI Centre, Beijing Normal University, Beijing, China
| |
Collapse
|
10
|
Bansal R, Singh R, Dutta TS, Dar ZA, Bajpai A. Indanone: a promising scaffold for new drug discovery against neurodegenerative disorders. Drug Discov Today 2024; 29:104063. [PMID: 38901670 DOI: 10.1016/j.drudis.2024.104063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/03/2024] [Accepted: 06/08/2024] [Indexed: 06/22/2024]
Abstract
Indanone is a versatile scaffold that has a number of pharmacological properties. The successful development and ensuing approval of indanone-derived donepezil as a drug of choice for Alzheimer's disease attracted significant scientific interest in this moiety. Indanones could act as small molecule chemical probes as they have strong affinity towards several critical enzymes associated with the pathophysiology of various neurological disorders. Inhibition of these enzymes elevates the levels of neuroprotective brain chemicals such as norepinephrine, serotonin and dopamine. Further, indanone derivatives are capable of modulating the activities of both monoamine oxidases (MAO-A and -B) and acetylcholinesterase (AChE), and thus could be useful in various neurodegenerative diseases. This review article presents a panoramic view of the research carried out on the indanone nucleus in the development of potential neuroprotective agents.
Collapse
Affiliation(s)
- Ranju Bansal
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India.
| | - Ranjit Singh
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Tuhin Shubra Dutta
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Zahid Ahmad Dar
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Ankit Bajpai
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| |
Collapse
|
11
|
Paiva I, Seguin J, Grgurina I, Singh AK, Cosquer B, Plassard D, Tzeplaeff L, Le Gras S, Cotellessa L, Decraene C, Gambi J, Alcala-Vida R, Eswaramoorthy M, Buée L, Cassel JC, Giacobini P, Blum D, Merienne K, Kundu TK, Boutillier AL. Dysregulated expression of cholesterol biosynthetic genes in Alzheimer's disease alters epigenomic signatures of hippocampal neurons. Neurobiol Dis 2024; 198:106538. [PMID: 38789057 DOI: 10.1016/j.nbd.2024.106538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024] Open
Abstract
Aging is the main risk factor of cognitive neurodegenerative diseases such as Alzheimer's disease, with epigenome alterations as a contributing factor. Here, we compared transcriptomic/epigenomic changes in the hippocampus, modified by aging and by tauopathy, an AD-related feature. We show that the cholesterol biosynthesis pathway is severely impaired in hippocampal neurons of tauopathic but not of aged mice pointing to vulnerability of these neurons in the disease. At the epigenomic level, histone hyperacetylation was observed at neuronal enhancers associated with glutamatergic regulations only in the tauopathy. Lastly, a treatment of tau mice with the CSP-TTK21 epi-drug that restored expression of key cholesterol biosynthesis genes counteracted hyperacetylation at neuronal enhancers and restored object memory. As acetyl-CoA is the primary substrate of both pathways, these data suggest that the rate of the cholesterol biosynthesis in hippocampal neurons may trigger epigenetic-driven changes, that may compromise the functions of hippocampal neurons in pathological conditions.
Collapse
Affiliation(s)
- Isabel Paiva
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France.
| | - Jonathan Seguin
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Iris Grgurina
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Akash Kumar Singh
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
| | - Brigitte Cosquer
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Damien Plassard
- University of Strasbourg, CNRS UMR7104, Inserm U1258 - GenomEast Platform - IGBMC - Institut de Génétique et de Biologie Moléculaire et Cellulaire, F-67404 Illkirch, France
| | - Laura Tzeplaeff
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Stephanie Le Gras
- University of Strasbourg, CNRS UMR7104, Inserm U1258 - GenomEast Platform - IGBMC - Institut de Génétique et de Biologie Moléculaire et Cellulaire, F-67404 Illkirch, France
| | - Ludovica Cotellessa
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, 59000 Lille, France
| | - Charles Decraene
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Johanne Gambi
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Rafael Alcala-Vida
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Muthusamy Eswaramoorthy
- Chemistry and Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France
| | - Jean-Christophe Cassel
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Paolo Giacobini
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, 59000 Lille, France
| | - David Blum
- University of Lille, Inserm, CHU Lille, UMR-S1172 LilNCog - Lille Neuroscience & Cognition, Lille, France; Alzheimer and Tauopathies, LabEx DISTALZ, France
| | - Karine Merienne
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, India
| | - Anne-Laurence Boutillier
- University of Strasbourg, Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France; CNRS, UMR7364 - Laboratoire de Neuroscience Cognitives et Adaptatives (LNCA), Strasbourg F-67000, France.
| |
Collapse
|
12
|
Hintermayer MA, Mendelson D, Byun JH. Consideration of T-Cell Profile in the Examination of Statin Efficacy in Inflammatory Diseases, Neurodegeneration, and Neurocognitive Performance. J Gerontol A Biol Sci Med Sci 2024; 79:glae156. [PMID: 38963798 PMCID: PMC11293203 DOI: 10.1093/gerona/glae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Indexed: 07/06/2024] Open
Abstract
Statins are a cornerstone in the medical management of cardiovascular disease, yet their efficacy varies greatly between individuals. In this commentary, we outline the evidence for the role of CD4+CD28null T-cell expansion as a critical moderator of the effects of statins in preventing cardiovascular events via the reduction of pathological inflammation. Given this relationship, we argue that T-cell profiles should be considered as a patient characteristic in clinical and preclinical studies examining statin efficacy in other age- and inflammation-related pathologies. We discuss the implications this may have for studies of statin use in numerous disease processes-notably, dementia and neurocognitive dysfunction-and the potential for T-cell profiles to be used as a prognosticator for statin efficacy in rheumatoid arthritis, Alzheimer's disease, and multiple sclerosis.
Collapse
Affiliation(s)
- Matthew A Hintermayer
- Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Québec, Canada
| | - Daniel Mendelson
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Québec, Canada
| | - Jae Hyun Byun
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Québec, Canada
| |
Collapse
|
13
|
Ha J, Kwon GE, Son Y, Jang SA, Cho SY, Park SJ, Kim H, Lee J, Lee J, Seo D, Lee M, Lee DY, Choi MH, Kim E. Cholesterol profiling reveals 7β-hydroxycholesterol as a pathologically relevant peripheral biomarker of Alzheimer's disease. Psychiatry Clin Neurosci 2024; 78:473-481. [PMID: 38923201 PMCID: PMC11488599 DOI: 10.1111/pcn.13706] [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: 11/27/2023] [Revised: 04/22/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
AIM Cholesterol homeostasis is associated with Alzheimer's disease (AD). Despite the multitude of cholesterol metabolites, little is known about which metabolites are directly involved in AD pathogenesis and can serve as its potential biomarkers. METHODS To identify "hit" metabolites, steroid profiling was conducted in mice with different age, diet, and genotype and also in humans with normal cognition, mild cognitive impairment, and AD using gas chromatography-mass spectrometry. Then, using one of the "hit" molecules (7β-hydroxycholesterol; OHC), molecular and histopathological experiment and behavioral testing were conducted in normal mice following its intracranial stereotaxic injection to see whether this molecule drives AD pathogenesis and causes cognitive impairment. RESULTS The serum levels of several metabolites, including 7β-OHC, were increased by aging in the 3xTg-AD unlike normal mice. Consistently, the levels of 7β-OHC were increased in the hairs of patients with AD and were correlated with clinical severity. We found that 7β-OHC directly affects AD-related pathophysiology; intrahippocampal injection of 7β-OHC induced astrocyte and microglial cell activation, increased the levels of pro-inflammatory cytokines (TNF-alpha, IL-1β, IL-6), and enhanced amyloidogenic pathway. Mice treated with 7β-OHC also exhibited deficits in memory and frontal/executive functions assessed by object recognition and 5-choice serial reaction time task, respectively. CONCLUSIONS Our results suggest that 7β-OHC could serve as a convenient, peripheral biomarker of AD. As directly involved in AD pathogenesis, 7β-OHC assay may help actualize personalized medicine in a way to identify an at-risk subgroup as a candidate population for statin-based AD treatment.
Collapse
Affiliation(s)
- Junghee Ha
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Go Eun Kwon
- Molecular Recognition Research CenterKorea Institute of Science and TechnologySeoulRepublic of Korea
| | - Yumi Son
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Graduate School of Medical Science, Brain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulRepublic of Korea
| | - Soo Ah Jang
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - So Yeon Cho
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Graduate School of Medical Science, Brain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulRepublic of Korea
| | - Soo Jin Park
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life SciencesSeoul National UniversitySeoulRepublic of Korea
| | - Hyunjeong Kim
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoulRepublic of Korea
| | - Jimin Lee
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Juseok Lee
- Department of MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Dongryul Seo
- Department of MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Myeongjee Lee
- Biostatistics Collaboration Unit, Department of Biomedical Systems InformaticsYonsei University College of MedicineSeoulKorea
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life SciencesSeoul National UniversitySeoulRepublic of Korea
| | - Man Ho Choi
- Molecular Recognition Research CenterKorea Institute of Science and TechnologySeoulRepublic of Korea
| | - Eosu Kim
- Department of Psychiatry, Laboratory for Alzheimer's Molecular Psychiatry, Institute of Behavioral Science in MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Graduate School of Medical Science, Brain Korea 21 PLUS Project for Medical ScienceYonsei University College of MedicineSeoulRepublic of Korea
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
14
|
Ahmed H, Wang Y, Griffiths WJ, Levey AI, Pikuleva I, Liang SH, Haider A. Brain cholesterol and Alzheimer's disease: challenges and opportunities in probe and drug development. Brain 2024; 147:1622-1635. [PMID: 38301270 PMCID: PMC11068113 DOI: 10.1093/brain/awae028] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 12/20/2023] [Accepted: 01/13/2024] [Indexed: 02/03/2024] Open
Abstract
Cholesterol homeostasis is impaired in Alzheimer's disease; however, attempts to modulate brain cholesterol biology have not translated into tangible clinical benefits for patients to date. Several recent milestone developments have substantially improved our understanding of how excess neuronal cholesterol contributes to the pathophysiology of Alzheimer's disease. Indeed, neuronal cholesterol was linked to the formation of amyloid-β and neurofibrillary tangles through molecular pathways that were recently delineated in mechanistic studies. Furthermore, remarkable advances in translational molecular imaging have now made it possible to probe cholesterol metabolism in the living human brain with PET, which is an important prerequisite for future clinical trials that target the brain cholesterol machinery in Alzheimer's disease patients-with the ultimate aim being to develop disease-modifying treatments. This work summarizes current concepts of how the biosynthesis, transport and clearance of brain cholesterol are affected in Alzheimer's disease. Further, current strategies to reverse these alterations by pharmacotherapy are critically discussed in the wake of emerging translational research tools that support the assessment of brain cholesterol biology not only in animal models but also in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Hazem Ahmed
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, 8093 Zurich, Switzerland
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - William J Griffiths
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Irina Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ahmed Haider
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
15
|
Chun MY, Chae W, Seo SW, Jang H, Yun J, Na DL, Kang D, Lee J, Hammers DB, Apostolova LG, Jang SI, Kim HJ. Effects of risk factors on the development and mortality of early- and late-onset dementia: an 11-year longitudinal nationwide population-based cohort study in South Korea. Alzheimers Res Ther 2024; 16:92. [PMID: 38664771 PMCID: PMC11044300 DOI: 10.1186/s13195-024-01436-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Early-onset dementia (EOD, onset age < 65) and late-onset dementia (LOD, onset age ≥ 65) exhibit distinct features. Understanding the risk factors for dementia development and mortality in EOD and LOD respectively is crucial for personalized care. While risk factors are known for LOD development and mortality, their impact on EOD remains unclear. We aimed to investigate how hypertension, diabetes mellitus, hyperlipidemia, atrial fibrillation, and osteoporosis influence the development and mortality of EOD and LOD, respectively. METHODS Using the Korean National Health Insurance Service (NHIS) database, we collected 546,709 dementia-free individuals and followed up for 11 years. In the two study groups, the Younger group (< 65 years old) and the Older group (≥ 65 years old), we applied Cox proportional hazard models to assess risk factors for development of EOD and LOD, respectively. Then, we assessed risk factors for mortality among EOD and LOD. RESULTS Diabetes mellitus and osteoporosis increased the risk of EOD and LOD development. Hypertension increased the risk of EOD, while atrial fibrillation increased the risk of LOD. Conversely, hyperlipidemia exhibited a protective effect against LOD development. Additionally, diabetes mellitus increased mortality in EOD and LOD. Hypertension and atrial fibrillation increased mortality in LOD, while hyperlipidemia decreased mortality in EOD and LOD. CONCLUSIONS Risk factors influencing dementia development and mortality differed in EOD and LOD. Targeted public health interventions addressing age-related risk factors may reduce dementia incidence and mortality.
Collapse
Affiliation(s)
- Min Young Chun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Neurology, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, South Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, 363 Dongbaekjukjeon-daero, Giheung-gu, , Yongin-si, Gyeonggi-do, 16995, South Korea
| | - Wonjeong Chae
- Office of Strategic Planning, Healthcare Policy and Strategy Task Force, Yonsei University Health System, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Neurology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, South Korea
| | - Jihwan Yun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
- Department of Neurology, Soonchunhyang University Bucheon Hospital, 170, Jomaru-ro, Wonmi-Gu, Bucheon-si, Gyeonggi-do, 14574, South Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Dongwoo Kang
- Department of Data Science, Hanmi Pharm. Co., Ltd, 14, Wiryeseong-daero, Songpa-gu, Seoul, South Korea
| | - Jungkuk Lee
- Department of Data Science, Hanmi Pharm. Co., Ltd, 14, Wiryeseong-daero, Songpa-gu, Seoul, South Korea
| | - Dustin B Hammers
- Department of Neurology, Indiana University School of Medicine, 355 W 16th St, Indianapolis, IN, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, 355 W 16th St, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine Indianapolis, 355W 16th St, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, 355W 16th St, Indianapolis, IN, USA
| | - Sung-In Jang
- Department of Preventive Medicine, College of Medicine, Yonsei University, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, South Korea.
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
- Department of Digital Health, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
- Department of Neurology, Indiana University School of Medicine, 355 W 16th St, Indianapolis, IN, USA.
| |
Collapse
|
16
|
Shin H, Ha WS, Kim J, Park SH, Han K, Baek MS. Association between migraine and the risk of vascular dementia: A nationwide longitudinal study in South Korea. PLoS One 2024; 19:e0300379. [PMID: 38630676 PMCID: PMC11023172 DOI: 10.1371/journal.pone.0300379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/26/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE We aimed to examine the potential association between migraine and vascular dementia (VaD) using a nationwide population database. BACKGROUND Migraine and VaD showed similar structural and functional changes in pathophysiology process and shared common risk factors, However, whether migraine prevalence increases VaD incidence remains controversial. METHODS This retrospective population-based cohort study used the medical records from the Korean National Health Insurance System database. Migraine (G43) was defined by using the Tenth Revision of the International Classification of Diseases code. More than two migraine diagnoses at least 3 months apart were defined as "chronic migraine". Cox proportional hazards model estimated hazard ratios (HRs) of VaD for group comparisons. RESULTS We included 212,836 patients with migraine and 5,863,348 individuals without migraine. During 10 years of follow-up, 3,914 (1.8%) and 60,258 (1.0%) patients with and without migraine, respectively, were newly diagnosed with VaD. After adjustment, patients with migraine showed a 1.21-fold higher risk of VaD than those without migraine (HR = 1.21; 95% confidence interval (CI): 1.17-1.25). Patients with chronic migraine showed a higher cumulative incidence of VaD than those with episodic migraine. The adjusted HR for the VaD incidence with migraine was higher in: (1) patients aged <65 years; (2) women; (3) patients without hypertension, diabetes, or atrial fibrillation; and (4) non-smokers. CONCLUSION Migraine is associated with an increased risk of VaD, particularly in chronic migraine patients. Incidence of VaD in the setting of migraine may have distinct pathophysiology from that of VaD with traditional cardiovascular risks.
Collapse
Affiliation(s)
- Hyomin Shin
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Woo Seok Ha
- Department of Neurology, Gangwon-do Wonju Medical Center, Wonju, South Korea
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jaeho Kim
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong-si, Gyeonggi-do, South Korea
| | - Sang Hyun Park
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, South Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, South Korea
| | - Min Seok Baek
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, South Korea
- Research Institute of Metabolism and Inflammation, Yonsei University Wonju College of Medicine, Wonju, South Korea
| |
Collapse
|
17
|
Sun M, Chen WM, Wu SY, Zhang J. Protective Effects Against Dementia Undergo Different Statin Type, Intensity, and Cumulative Dose in Older Adult Type 2 Diabetes Mellitus Patients. J Am Med Dir Assoc 2024; 25:470-479.e1. [PMID: 38128583 DOI: 10.1016/j.jamda.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVES This study investigated the relationship between statin use and dementia risk in older adults with type 2 diabetes (T2DM). It also assessed the impact of various statin types, dosage intensity, and cumulative doses on dementia risk. DESIGN Employing the inverse probability of treatment weighting (IPTW) Cox hazards model, this research explored the influence of statin utilization on dementia incidence. SETTING AND PARTICIPANTS The study included older adult T2DM patients aged 60 years or older who received statins (case group) and those who did not (control group) during the follow-up period. METHODS The IPTW Cox hazards model quantified the association between statin use and dementia incidence. Subgroup analyses investigated different statin types, usage intensity, and cumulative dose-dependent relationships with dementia risk, measured by adjusted hazard ratios (aHRs) with corresponding 95% CIs. RESULTS Statin users experienced a significant reduction in dementia risk (aHR: 0.47, 95% CI: 0.46-0.48). Subgroup analysis using IPTW Cox regression revealed varying dementia incidence reductions among users of different statin types, with aHRs (95% CIs) ranging from 0.09 to 0.69. Multivariate analyses unveiled a dose-dependent relationship, showing reduced dementia incidence based on cumulative defined daily doses (cDDDs) per year. The corresponding aHRs (95% CIs) were 0.20 to 0.72 across quartiles 4 to 1 of cDDD-years, with a significant trend (P < .001). The optimal daily statin use was 0.88 defined daily doses (DDDs), associated with the lowest dementia risk. CONCLUSIONS AND IMPLICATIONS Statins significantly reduced dementia risk in older adult T2DM patients. Higher cumulative defined daily doses (cDDD-years) were linked to more substantial risk reductions. This research underscores the clinical benefits of statin use in preventing dementia in this population and calls for further investigation into the underlying mechanisms. It also raises the possibility of influencing policy decisions to manage dementia risk in this vulnerable group.
Collapse
Affiliation(s)
- Mingyang Sun
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Wan-Ming Chen
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan; Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan
| | - Szu-Yuan Wu
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei, Taiwan; Artificial Intelligence Development Center, Fu Jen Catholic University, Taipei, Taiwan; Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan; Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Division of Radiation Oncology, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Department of Healthcare Administration, College of Medical and Health Science, Asia University, Taichung, Taiwan; Cancer Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan; Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Jiaqiang Zhang
- Department of Anesthesiology and Perioperative Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
| |
Collapse
|
18
|
Pappolla MA, Refolo L, Sambamurti K, Zambon D, Duff K. Hypercholesterolemia and Alzheimer's Disease: Unraveling the Connection and Assessing the Efficacy of Lipid-Lowering Therapies. J Alzheimers Dis 2024; 101:S371-S393. [PMID: 39422957 DOI: 10.3233/jad-240388] [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: 10/19/2024]
Abstract
This article examines the relationship between cholesterol levels and Alzheimer's disease (AD), beginning with the early observation that individuals who died from heart attacks often had brain amyloid deposition. Subsequent animal model research proved that high cholesterol could hasten amyloid accumulation. In contrast, cholesterol-lowering treatments appeared to counteract this effect. Human autopsy studies reinforced the cholesterol-AD connection, revealing that higher cholesterol levels during midlife significantly correlated with higher brain amyloid pathology. This effect was especially pronounced in individuals aged 40 to 55. Epidemiological data supported animal research and human tissue observations and suggested that managing cholesterol levels in midlife could reduce the risk of developing AD. We analyze the main observational studies and clinical trials on the efficacy of statins. While observational data often suggest a potential protective effect against AD, clinical trials have not consistently shown benefit. The failure of these trials to demonstrate a clear advantage is partially attributed to multiple factors, including the timing of statin therapy, the type of statin and the appropriate selection of patients for treatment. Many studies failed to target individuals who might benefit most from early intervention, such as high-risk patients like APOE4 carriers. The review addresses how cholesterol is implicated in AD through various biological pathways, the potential preventive role of cholesterol management as suggested by observational studies, and the difficulties encountered in clinical trials, particularly related to statin use. The paper highlights the need to explore alternate therapeutic targets and mechanisms that escape statin intervention.
Collapse
Affiliation(s)
- Miguel A Pappolla
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lorenzo Refolo
- Translational Research Branch, Division of Neuroscience, Bethesda, MD, USA
| | - Kumar Sambamurti
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel Zambon
- Universitat Internacional de Catalunya, Barcelona, Spain
| | - Karen Duff
- Karen Duff, UK Dementia Research Institute at University College London, London, UK
| |
Collapse
|
19
|
Zhou X, Wu X, Wang R, Han L, Li H, Zhao W. Mechanisms of 3-Hydroxyl 3-Methylglutaryl CoA Reductase in Alzheimer's Disease. Int J Mol Sci 2023; 25:170. [PMID: 38203341 PMCID: PMC10778631 DOI: 10.3390/ijms25010170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide and has a high incidence in the elderly. Unfortunately, there is no effective therapy for AD owing to its complicated pathogenesis. However, the development of lipid-lowering anti-inflammatory drugs has heralded a new era in the treatment of Alzheimer's disease. Several studies in recent years have shown that lipid metabolic dysregulation and neuroinflammation are associated with the pathogenesis of AD. 3-Hydroxyl 3-methylglutaryl CoA reductase (HMGCR) is a rate-limiting enzyme in cholesterol synthesis that plays a key role in cholesterol metabolism. HMGCR inhibitors, known as statins, have changed from being solely lipid-lowering agents to neuroprotective compounds because of their effects on lipid levels and inflammation. In this review, we first summarize the main regulatory mechanism of HMGCR affecting cholesterol biosynthesis. We also discuss the pathogenesis of AD induced by HMGCR, including disordered lipid metabolism, oxidative stress, inflammation, microglial proliferation, and amyloid-β (Aβ) deposition. Subsequently, we explain the possibility of HMGCR as a potential target for AD treatment. Statins-based AD treatment is an ascent field and currently quite controversial; therefore, we also elaborate on the current application prospects and limitations of statins in AD treatment.
Collapse
Affiliation(s)
- Xun Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
- Department of Endocrinology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China;
| | - Xiaolang Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Rui Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Lu Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Huilin Li
- Department of Endocrinology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China;
| | - Wei Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| |
Collapse
|
20
|
Wang C, Hou J, Zhang M, Zheng Y, Ye H, Qi Y, Guo L, Hu Y. Effects of HSYA on serum and brain cholesterol levels in AD rats based on quantitative proteomics. Int J Neurosci 2023; 133:1411-1423. [PMID: 35633062 DOI: 10.1080/00207454.2022.2082964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/17/2022] [Indexed: 10/18/2022]
Abstract
Backgroud: Hydroxysafflor yellow A (HSYA) has a certain improvement effect on Alzheimer's disease (AD) rats, but its specific mechanism is still unclear. The purpose of this study was to observe the regulatory effect of HSYA on learning and memory ability of AD rats induced by Aβ1-42.Materials and methods: Morris water maze test was used to evaluate the effect of HSYA on the learning and memory ability of AD model rats. To explore the effective targets and potential molecular mechanisms of HSYA in AD treatment based on quantitative proteomics.Results: Through the Morris water maze experiment, we found that after HSYA treatment, the learning ability of rats in the model group has been significantly improved. Quantitative proteomics results showed that among the 11 common differential proteins between the "model/sham operation" comparison group and the "HSYA treatment/model" comparison group, the cholesterol synthesis rate-limiting enzyme mevalonate decarboxylase (Mvd) Western Blot results are consistent with the results of quantitative proteomics analysis. We found that HSYA can inhibit the expression of BACE protein in hippocampus of AD rats and decrease the level of Aβ1-42. Besides, HSYA could also reduce cholesterol levels in serum and hippocampus.Conclusion: In summary, HSYA can effectively improve learning and memory disorders in AD rats, and exert neuroprotective effects by effectively controlling serum and brain cholesterol to down-regulate the expression of BACE and thus reduce the content of Aβ1-42.
Collapse
Affiliation(s)
- Chunhui Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, P.R. China
| | - Jiawei Hou
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, P.R. China
- School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mengyu Zhang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, P.R. China
| | - Yanjie Zheng
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, P.R. China
| | - Hongxia Ye
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, P.R. China
| | - Yanqiang Qi
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, P.R. China
| | - Li Guo
- State Key Laboratory of Biobased Material and Green Papermaking, School of Food Sciences and Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China
| | - Yanli Hu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, P.R. China
| |
Collapse
|
21
|
Hu ZL, Yuan YQ, Tong Z, Liao MQ, Yuan SL, Jian Y, Yang JL, Liu WF. Reexamining the Causes and Effects of Cholesterol Deposition in the Brains of Patients with Alzheimer's Disease. Mol Neurobiol 2023; 60:6852-6868. [PMID: 37507575 DOI: 10.1007/s12035-023-03529-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system. Numerous studies have shown that imbalances in cholesterol homeostasis in the brains of AD patients precede the onset of clinical symptoms. In addition, cholesterol deposition has been observed in the brains of AD patients even though peripheral cholesterol does not enter the brain through the blood‒brain barrier (BBB). Studies have demonstrated that cholesterol metabolism in the brain is associated with many pathological conditions, such as amyloid beta (Aβ) production, Tau protein phosphorylation, oxidative stress, and inflammation. In 2022, some scholars put forward a new hypothesis of AD: the disease involves lipid invasion and its exacerbation of the abnormal metabolism of cholesterol in the brain. In this review, by discussing the latest research progress, the causes and effects of cholesterol retention in the brains of AD patients are analyzed and discussed. Additionally, the possible mechanism through which AD may be improved by targeting cholesterol is described. Finally, we propose that improving the impairments in cholesterol removal observed in the brains of AD patients, instead of further reducing the already impaired cholesterol synthesis in the brain, may be the key to preventing cholesterol deposition and improving the corresponding pathological symptoms.
Collapse
Affiliation(s)
- Ze-Lin Hu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Yang-Qi Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zhen Tong
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Mei-Qing Liao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Shun-Ling Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Ye Jian
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Jia-Lun Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Wen-Feng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
22
|
Essayan-Perez S, Südhof TC. Neuronal γ-secretase regulates lipid metabolism, linking cholesterol to synaptic dysfunction in Alzheimer's disease. Neuron 2023; 111:3176-3194.e7. [PMID: 37543038 PMCID: PMC10592349 DOI: 10.1016/j.neuron.2023.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/16/2023] [Accepted: 07/10/2023] [Indexed: 08/07/2023]
Abstract
Presenilin mutations that alter γ-secretase activity cause familial Alzheimer's disease (AD), whereas ApoE4, an apolipoprotein for cholesterol transport, predisposes to sporadic AD. Both sporadic and familial AD feature synaptic dysfunction. Whether γ-secretase is involved in cholesterol metabolism and whether such involvement impacts synaptic function remains unknown. Here, we show that in human neurons, chronic pharmacological or genetic suppression of γ-secretase increases synapse numbers but decreases synaptic transmission by lowering the presynaptic release probability without altering dendritic or axonal arborizations. In search of a mechanism underlying these synaptic impairments, we discovered that chronic γ-secretase suppression robustly decreases cholesterol levels in neurons but not in glia, which in turn stimulates neuron-specific cholesterol-synthesis gene expression. Suppression of cholesterol levels by HMG-CoA reductase inhibitors (statins) impaired synaptic function similar to γ-secretase inhibition. Thus, γ-secretase enables synaptic function by maintaining cholesterol levels, whereas the chronic suppression of γ-secretase impairs synapses by lowering cholesterol levels.
Collapse
Affiliation(s)
- Sofia Essayan-Perez
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
23
|
Valenza M, Birolini G, Cattaneo E. The translational potential of cholesterol-based therapies for neurological disease. Nat Rev Neurol 2023; 19:583-598. [PMID: 37644213 DOI: 10.1038/s41582-023-00864-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
Cholesterol is an important metabolite and membrane component and is enriched in the brain owing to its role in neuronal maturation and function. In the adult brain, cholesterol is produced locally, predominantly by astrocytes. When cholesterol has been used, recycled and catabolized, the derivatives are excreted across the blood-brain barrier. Abnormalities in any of these steps can lead to neurological dysfunction. Here, we examine how precise interactions between cholesterol production and its use and catabolism in neurons ensures cholesterol homeostasis to support brain function. As an example of a neurological disease associated with cholesterol dyshomeostasis, we summarize evidence from animal models of Huntington disease (HD), which demonstrate a marked reduction in cholesterol biosynthesis with clinically relevant consequences for synaptic activity and cognition. In addition, we examine the relationship between cholesterol loss in the brain and cognitive decline in ageing. We then present emerging therapeutic strategies to restore cholesterol homeostasis, focusing on evidence from HD mouse models.
Collapse
Affiliation(s)
- Marta Valenza
- Department of Biosciences, University of Milan, Milan, Italy.
- Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy.
| | - Giulia Birolini
- Department of Biosciences, University of Milan, Milan, Italy
- Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Elena Cattaneo
- Department of Biosciences, University of Milan, Milan, Italy.
- Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy.
| |
Collapse
|
24
|
Rudajev V, Novotny J. Cholesterol-dependent amyloid β production: space for multifarious interactions between amyloid precursor protein, secretases, and cholesterol. Cell Biosci 2023; 13:171. [PMID: 37705117 PMCID: PMC10500844 DOI: 10.1186/s13578-023-01127-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
Amyloid β is considered a key player in the development and progression of Alzheimer's disease (AD). Many studies investigating the effect of statins on lowering cholesterol suggest that there may be a link between cholesterol levels and AD pathology. Since cholesterol is one of the most abundant lipid molecules, especially in brain tissue, it affects most membrane-related processes, including the formation of the most dangerous form of amyloid β, Aβ42. The entire Aβ production system, which includes the amyloid precursor protein (APP), β-secretase, and the complex of γ-secretase, is highly dependent on membrane cholesterol content. Moreover, cholesterol can affect amyloidogenesis in many ways. Cholesterol influences the stability and activity of secretases, but also dictates their partitioning into specific cellular compartments and cholesterol-enriched lipid rafts, where the amyloidogenic machinery is predominantly localized. The most complicated relationships have been found in the interaction between cholesterol and APP, where cholesterol affects not only APP localization but also the precise character of APP dimerization and APP processing by γ-secretase, which is important for the production of Aβ of different lengths. In this review, we describe the intricate web of interdependence between cellular cholesterol levels, cholesterol membrane distribution, and cholesterol-dependent production of Aβ, the major player in AD.
Collapse
Affiliation(s)
- Vladimir Rudajev
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jiri Novotny
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
25
|
Thong EHE, Quek EJW, Loo JH, Yun CY, Teo YN, Teo YH, Leow AST, Li TYW, Sharma VK, Tan BYQ, Yeo LLL, Chong YF, Chan MY, Sia CH. Acute Myocardial Infarction and Risk of Cognitive Impairment and Dementia: A Review. BIOLOGY 2023; 12:1154. [PMID: 37627038 PMCID: PMC10452707 DOI: 10.3390/biology12081154] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/05/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Cognitive impairment (CI) shares common cardiovascular risk factors with acute myocardial infarction (AMI), and is increasingly prevalent in our ageing population. Whilst AMI is associated with increased rates of CI, CI remains underreported and infrequently identified in patients with AMI. In this review, we discuss the evidence surrounding AMI and its links to dementia and CI, including pathophysiology, risk factors, management and interventions. Vascular dysregulation plays a major role in CI, with atherosclerosis, platelet activation, microinfarcts and perivascular inflammation resulting in neurovascular unit dysfunction, disordered homeostasis and a dysfunctional neurohormonal response. This subsequently affects perfusion pressure, resulting in enlarged periventricular spaces and hippocampal sclerosis. The increased platelet activation seen in coronary artery disease (CAD) can also result in inflammation and amyloid-β protein deposition which is associated with Alzheimer's Dementia. Post-AMI, reduced blood pressure and reduced left ventricular ejection fraction can cause chronic cerebral hypoperfusion, cerebral infarction and failure of normal circulatory autoregulatory mechanisms. Patients who undergo coronary revascularization (percutaneous coronary intervention or bypass surgery) are at increased risk for post-procedure cognitive impairment, though whether this is related to the intervention itself or underlying cardiovascular risk factors is debated. Mortality rates are higher in dementia patients with AMI, and post-AMI CI is more prevalent in the elderly and in patients with post-AMI heart failure. Medical management (antiplatelet, statin, renin-angiotensin system inhibitors, cardiac rehabilitation) can reduce the risk of post-AMI CI; however, beta-blockers may be associated with functional decline in patients with existing CI. The early identification of those with dementia or CI who present with AMI is important, as subsequent tailoring of management strategies can potentially improve outcomes as well as guide prognosis.
Collapse
Affiliation(s)
- Elizabeth Hui En Thong
- Internal Medicine Residency, National University Health System, Singapore 119074, Singapore; (E.H.E.T.); (Y.H.T.); (A.S.T.L.)
| | - Ethan J. W. Quek
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
| | - Jing Hong Loo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
| | - Choi-Ying Yun
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| | - Yao Neng Teo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
| | - Yao Hao Teo
- Internal Medicine Residency, National University Health System, Singapore 119074, Singapore; (E.H.E.T.); (Y.H.T.); (A.S.T.L.)
| | - Aloysius S. T. Leow
- Internal Medicine Residency, National University Health System, Singapore 119074, Singapore; (E.H.E.T.); (Y.H.T.); (A.S.T.L.)
| | - Tony Y. W. Li
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| | - Vijay K. Sharma
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Benjamin Y. Q. Tan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Leonard L. L. Yeo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Yao Feng Chong
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Mark Y. Chan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| | - Ching-Hui Sia
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| |
Collapse
|
26
|
Min JO, Ho HA, Lee W, Jung BC, Park SJ, Kim S, Lee SJ. Statins suppress cell-to-cell propagation of α-synuclein by lowering cholesterol. Cell Death Dis 2023; 14:474. [PMID: 37500624 PMCID: PMC10374525 DOI: 10.1038/s41419-023-05977-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 06/27/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023]
Abstract
Cell-to-cell propagation of protein aggregates has been implicated in the progression of neurodegenerative diseases. However, the underlying mechanism and modulators of this process are not fully understood. Here, we screened a small-molecule library in a search for agents that suppress the propagation of α-synuclein and mutant huntingtin (mHtt). These screens yielded several molecules, some of which were effective against both α-synuclein and mHtt. Among these molecules, we focused on simvastatin and pravastatin. Simvastatin administration in a transgenic model of synucleinopathy effectively ameliorated behavioral deficits and α-synuclein accumulation, whereas pravastatin had no effect. Because only simvastatin enters the brain effectively, these results suggest that inhibition of brain cholesterol synthesis is important in simvastatin effects. In cultured cells, accumulation of intracellular cholesterol, induced by genetic ablation of the NPC1 gene or by pharmacological treatment with U18666A, increased α-synuclein aggregation and secretion. In contrast, lowering cholesterol using methyl-β-cyclodextrin or statins reversed α-synuclein aggregation and secretion in NPC1-knockout cells. Consistent with these observations, feeding a high-fat diet aggravated α-synuclein pathology and behavioral deficits in the preformed fibril-injected mouse model, an effect that was also reversed by simvastatin administration. These results suggest that statins suppress propagation of protein aggregates by lowering cholesterol in the brain.
Collapse
Affiliation(s)
- Joo-Ok Min
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hoang-Anh Ho
- Interdisciplinary Program in Neuroscience, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Wonjae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Neuramedy Co. Ltd, Seoul, Republic of Korea
| | - Byung Chul Jung
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, CA, USA
| | - Sung Jun Park
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | | | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Neuramedy Co. Ltd, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Tang B, Li X, Wang Y, Sjölander A, Johnell K, Thambisetty M, Ferrucci L, Reynolds CA, Finkel D, Jylhävä J, Pedersen NL, Hägg S. Longitudinal associations between use of antihypertensive, antidiabetic, and lipid-lowering medications and biological aging. GeroScience 2023:10.1007/s11357-023-00784-8. [PMID: 37032369 PMCID: PMC10400489 DOI: 10.1007/s11357-023-00784-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/26/2023] [Indexed: 04/11/2023] Open
Abstract
Aging is a major risk factor for many chronic diseases. This study aimed to examine the effects of antihypertensive, lipid-lowering, and antidiabetic drugs on biological aging. We included 672 participants and 2746 repeated measurements from the Swedish Adoption/Twin Study of Aging. Self-reported medicine uses were categorized into antidiabetic, antihypertensive, and lipid-lowering drugs. A total of 12 biomarkers for biological aging (BA biomarkers) were included as outcomes. Conditional generalized estimating equations were applied conditioning on individuals to estimate the drug effect on BA biomarker level within the same person when using or not using the drug. Chronological age, body mass index, smoking status, number of multiple medication uses, blood pressure, blood glucose level, and apoB/apoA ratio were adjusted for as covariates in the model. Overall, using antihypertensive drugs was associated with a decrease in one DNA-methylation age (PCGrimAge: beta = - 0.39, 95%CI = - 0.67 to - 0.12). When looking into drug subcategories, calcium channel blockers (CCBs) were associated with a decrease in several DNA-methylation ages (PCHorvathAge beta = - 1.28, 95%CI = - 2.34 to - 0.21; PCSkin&bloodAge beta = - 1.34, 95%CI = - 2.61 to - 0.07; PCPhenoAge beta = - 1.74, 95%CI = - 2.58 to - 0.89; PCGrimAge beta = - 0.57, 95%CI = - 0.96 to - 0.17) and in functional biological ages (functional age index beta = - 2.18, 95%CI = - 3.65 to - 0.71; frailty index beta = - 1.31, 95%CI = - 2.43 to - 0.18). However, the results within other drug subcategories were inconsistent. Calcium channel blockers may decrease biological aging captured by the BA biomarkers measured at epigenetic and functional level. Future studies are warranted to confirm these effects and understand the underlying biological mechanisms.
Collapse
Affiliation(s)
- Bowen Tang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Xia Li
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Yunzhang Wang
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Solna, Sweden
| | - Arvid Sjölander
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Kristina Johnell
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Madhav Thambisetty
- Brain Aging and Behavior Section, National Institute on Aging, Baltimore, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, National Institute on Aging, Baltimore, USA
| | | | - Deborah Finkel
- Aging Research Network-Jönköping (ARN-J), School of Health and Welfare, Jönköping University, Jönköping, Sweden
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, USA
| | - Juulia Jylhävä
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
- Faculty of Social Sciences (Health Sciences) and Gerontology Research Center (GEREC), University of Tampere, Tampere, Finland
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
28
|
Rahman AA, Michaud J, Dell'Aniello S, Moodie EEM, Brophy JM, Durand M, Guertin JR, Boivin JF, Renoux C. Oral Anticoagulants and the Risk of Dementia in Patients With Nonvalvular Atrial Fibrillation: A Population-Based Cohort Study. Neurology 2023; 100:e1309-e1320. [PMID: 36581462 PMCID: PMC10033170 DOI: 10.1212/wnl.0000000000206748] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 11/15/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Nonvalvular atrial fibrillation (NVAF) is associated with an increased risk of dementia. Oral anticoagulants (OACs) are essential for stroke prevention in NVAF, and studies have shown a possible protective effect on dementia. However, findings have been inconsistent and hampered by methodological limitations. Thus, we assessed whether the use of OACs is associated with a decreased incidence of dementia in patients with NVAF. In addition, we explored the impact of the cumulative duration of OAC use on the incidence of dementia. METHODS Using the UK Clinical Practice Research Datalink, we formed a cohort of all patients aged 50 years or older with an incident diagnosis of NVAF between 1988 and 2017 and no prior OAC use, with a follow-up until 2019. Patients were considered unexposed until 6 months after their first OAC prescription for latency considerations and exposed thereafter until the end of follow-up. We used time-dependent Cox regression models to estimate hazard ratios (HRs), adjusted for 54 covariates, with 95% CIs for dementia associated with OAC use, compared with nonuse. We also assessed whether the risk varied with the cumulative duration of OAC use, compared with nonuse, by comparing prespecified exposure categories defined in a time-varying manner and by modeling the HR using a restricted cubic spline. RESULTS The cohort included 142,227 patients with NVAF, with 8,023 cases of dementia over 662,667 person-years of follow-up (incidence rate 12.1, 95% CI 11.9-12.4 per 1,000 person-years). OAC use was associated with a decreased risk of dementia (HR 0.88, 95% CI 0.84-0.92) compared with nonuse. A restricted cubic spline also indicated a decreased risk of dementia, reaching a low at approximately 1.5 years of cumulative OAC use and stabilizing thereafter. Moreover, OAC use decreased the risk in patients aged 75 years and older (HR 0.84, 95% CI 0.80-0.89), but not in younger patients (HR 0.99, 95% CI 0.90-1.10). DISCUSSION In patients with incident NVAF, OACs were associated with a decreased risk of dementia, particularly in elderly individuals. This warrants consideration when weighing the risks and benefits of anticoagulation in this population. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that in patients with NVAF, OAC use (vs nonuse) is associated with a decreased risk of dementia.
Collapse
Affiliation(s)
- Alvi A Rahman
- From the Departments of Epidemiology, Biostatistics and Occupational Health (A.R., E.E.M.M., J.M.B., J.-F.B., C.R.), Medicine (J.M.B.), and Neurology and Neurosurgery (C.R.), McGill University, Montreal; Centre for Clinical Epidemiology (A.R., J.M., S.D., J.-F.B., C.R.), Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Department of Medicine (M.D.), Université de Montréal; Centre de recherche du Centre Hospitalier de l'Université de Montréal (M.D.); and Département de Médecine Sociale et Préventive (J.R.G.), Faculté de médecine, Université Laval, Montreal, Canada
| | - Jonathan Michaud
- From the Departments of Epidemiology, Biostatistics and Occupational Health (A.R., E.E.M.M., J.M.B., J.-F.B., C.R.), Medicine (J.M.B.), and Neurology and Neurosurgery (C.R.), McGill University, Montreal; Centre for Clinical Epidemiology (A.R., J.M., S.D., J.-F.B., C.R.), Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Department of Medicine (M.D.), Université de Montréal; Centre de recherche du Centre Hospitalier de l'Université de Montréal (M.D.); and Département de Médecine Sociale et Préventive (J.R.G.), Faculté de médecine, Université Laval, Montreal, Canada
| | - Sophie Dell'Aniello
- From the Departments of Epidemiology, Biostatistics and Occupational Health (A.R., E.E.M.M., J.M.B., J.-F.B., C.R.), Medicine (J.M.B.), and Neurology and Neurosurgery (C.R.), McGill University, Montreal; Centre for Clinical Epidemiology (A.R., J.M., S.D., J.-F.B., C.R.), Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Department of Medicine (M.D.), Université de Montréal; Centre de recherche du Centre Hospitalier de l'Université de Montréal (M.D.); and Département de Médecine Sociale et Préventive (J.R.G.), Faculté de médecine, Université Laval, Montreal, Canada
| | - Erica E M Moodie
- From the Departments of Epidemiology, Biostatistics and Occupational Health (A.R., E.E.M.M., J.M.B., J.-F.B., C.R.), Medicine (J.M.B.), and Neurology and Neurosurgery (C.R.), McGill University, Montreal; Centre for Clinical Epidemiology (A.R., J.M., S.D., J.-F.B., C.R.), Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Department of Medicine (M.D.), Université de Montréal; Centre de recherche du Centre Hospitalier de l'Université de Montréal (M.D.); and Département de Médecine Sociale et Préventive (J.R.G.), Faculté de médecine, Université Laval, Montreal, Canada
| | - James M Brophy
- From the Departments of Epidemiology, Biostatistics and Occupational Health (A.R., E.E.M.M., J.M.B., J.-F.B., C.R.), Medicine (J.M.B.), and Neurology and Neurosurgery (C.R.), McGill University, Montreal; Centre for Clinical Epidemiology (A.R., J.M., S.D., J.-F.B., C.R.), Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Department of Medicine (M.D.), Université de Montréal; Centre de recherche du Centre Hospitalier de l'Université de Montréal (M.D.); and Département de Médecine Sociale et Préventive (J.R.G.), Faculté de médecine, Université Laval, Montreal, Canada
| | - Madeleine Durand
- From the Departments of Epidemiology, Biostatistics and Occupational Health (A.R., E.E.M.M., J.M.B., J.-F.B., C.R.), Medicine (J.M.B.), and Neurology and Neurosurgery (C.R.), McGill University, Montreal; Centre for Clinical Epidemiology (A.R., J.M., S.D., J.-F.B., C.R.), Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Department of Medicine (M.D.), Université de Montréal; Centre de recherche du Centre Hospitalier de l'Université de Montréal (M.D.); and Département de Médecine Sociale et Préventive (J.R.G.), Faculté de médecine, Université Laval, Montreal, Canada
| | - Jason R Guertin
- From the Departments of Epidemiology, Biostatistics and Occupational Health (A.R., E.E.M.M., J.M.B., J.-F.B., C.R.), Medicine (J.M.B.), and Neurology and Neurosurgery (C.R.), McGill University, Montreal; Centre for Clinical Epidemiology (A.R., J.M., S.D., J.-F.B., C.R.), Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Department of Medicine (M.D.), Université de Montréal; Centre de recherche du Centre Hospitalier de l'Université de Montréal (M.D.); and Département de Médecine Sociale et Préventive (J.R.G.), Faculté de médecine, Université Laval, Montreal, Canada
| | - Jean-François Boivin
- From the Departments of Epidemiology, Biostatistics and Occupational Health (A.R., E.E.M.M., J.M.B., J.-F.B., C.R.), Medicine (J.M.B.), and Neurology and Neurosurgery (C.R.), McGill University, Montreal; Centre for Clinical Epidemiology (A.R., J.M., S.D., J.-F.B., C.R.), Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Department of Medicine (M.D.), Université de Montréal; Centre de recherche du Centre Hospitalier de l'Université de Montréal (M.D.); and Département de Médecine Sociale et Préventive (J.R.G.), Faculté de médecine, Université Laval, Montreal, Canada
| | - Christel Renoux
- From the Departments of Epidemiology, Biostatistics and Occupational Health (A.R., E.E.M.M., J.M.B., J.-F.B., C.R.), Medicine (J.M.B.), and Neurology and Neurosurgery (C.R.), McGill University, Montreal; Centre for Clinical Epidemiology (A.R., J.M., S.D., J.-F.B., C.R.), Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada; Department of Medicine (M.D.), Université de Montréal; Centre de recherche du Centre Hospitalier de l'Université de Montréal (M.D.); and Département de Médecine Sociale et Préventive (J.R.G.), Faculté de médecine, Université Laval, Montreal, Canada.
| |
Collapse
|
29
|
Nechyporenko A, Tedla YG, Korcarz C, Tattersall MC, Greenland P, Gepner AD. Association of statin therapy with progression of carotid arterial stiffness: the Multi-Ethnic Study of Atherosclerosis (MESA). Hypertens Res 2023; 46:679-687. [PMID: 36434289 DOI: 10.1038/s41440-022-01095-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/30/2022] [Accepted: 09/19/2022] [Indexed: 11/27/2022]
Abstract
Arterial stiffness progresses with age and is a predictor of adverse cardiovascular disease events. Studies examining associations of statin therapy with arterial stiffness have yielded mixed results. Associations between the duration and intensity of statin therapy and arterial stiffness have not been studied in a prospective multiethnic cohort. MESA participants (n = 1242) with statin medication use data at each exam (1-5) and who had undergone B-mode carotid ultrasound at baseline and at Exam 5 after (mean ± [SD]) 9.4 ± 0.5 years were analyzed. Carotid arterial stiffness was measured using the distensibility coefficient (DC) and Young's elastic modulus (YEM). Linear regression models were used to evaluate associations between DC and YEM and statin treatment duration and intensity. At baseline, participants were 66.5 ± 8.1 years old, 41% female, 36% White, 30% African American, 14% Chinese American, and 20% Hispanic. The mean baseline low-density lipoprotein cholesterol (LDL-C) was 149.5 ± 14.5 mg/dL. After adjusting for age, sex, race/ethnicity, and CVD risk factors, the percent changes in DC and YEM were found to not be significantly different in individuals on statin therapy at any combination of visits (1-4) compared to participants never on statin therapy (all p > 0.32). There were also no differences in the percent change in DC and YEM based on statin therapy intensity by quartile (all p > 0.14) over the 10-year follow-up period. Based on the aforementioned results, statin therapy was not associated with changes in carotid artery stiffness over nearly a decade of follow-up regardless of therapy duration or intensity.
Collapse
Affiliation(s)
- Anatoliy Nechyporenko
- Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Yacob G Tedla
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Claudia Korcarz
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | | | - Philip Greenland
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Adam D Gepner
- Department of Medicine, University of Wisconsin, Madison, WI, USA. .,William S. Middleton VA Hospital, Madison, WI, USA.
| |
Collapse
|
30
|
A Review on Phyto-Therapeutic Approaches in Alzheimer's Disease. J Funct Biomater 2023; 14:jfb14010050. [PMID: 36662097 PMCID: PMC9861153 DOI: 10.3390/jfb14010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/29/2022] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
Neurodegenerative diseases occur due to progressive and sometimes irreversible loss of function and death of nerve cells. A great deal of effort is being made to understand the pathogenesis of neurodegenerative diseases. In particular, the prevalence of Alzheimer's disease (AD) is quite high, and only symptomatic therapy is available due to the absence of radical treatment. The aim of this review is to try to elucidate the general pathogenesis of AD, to provide information about the limit points of symptomatic treatment approaches, and to emphasize the potential neurologic effects of phytocompounds as new tools as therapeutic agents for disease prevention, retardation, and therapy. This survey also covers the notable properties of herbal compounds such as their effects on the inhibition of an enzyme called acetylcholinesterase, which has significant value in the treatment of AD. It has been proven that phytopharmaceuticals have long-term effects that could protect nervous system health, eliminate inflammatory responses, improve cognitive damage, provide anti-aging effects in the natural aging process, and alleviate dementia sequelae. Herbal-based therapeutic agents can afford many advantages and can be used as potentially as new-generation therapeutics or complementary agents with high compliance, fewer adverse effects, and lower cost in comparison to the traditional pharmaceutical agents in the fight against AD.
Collapse
|
31
|
Murphy C, Dyer AH, Lawlor B, Kennelly SP. What is the impact of ongoing statin use on cognitive decline and dementia progression in older adults with mild-moderate Alzheimer disease? PLoS One 2023; 18:e0285529. [PMID: 37167234 PMCID: PMC10174559 DOI: 10.1371/journal.pone.0285529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/25/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND In addition to the clear cardiovascular benefit, there has been renewed interest in the potential of statins in the prevention of cognitive impairment and dementia in older adults. However, whether ongoing statin use can delay cognitive decline or dementia progression in those with established Alzheimer dementia, is unclear. METHODS Using data from NILVAD, we analysed the association between ongoing statin use and cognitive decline (Alzheimer Disease Assessment Scale-Cognitive Subsection [ADAS-Cog])/dementia progression (Clinical Dementia Rating Scale [CDR-Sb]/Disability Assessment for Dementia [DAD]) over 18 months in older adults with mild-moderate AD. Additionally, we assessed the association between ongoing statin use and adverse events in mild-moderate AD. RESULTS Over one-third (34.9%) of 510 older adults with mild-moderate AD (aged: 72.9 years; 61.9% female) used a statin for the 18-month study duration. Statin use was not associated with the rate of cognitive decline (β: -0.67; 95% CI: -1.71, 0.36, p = 0.20) or dementia progression (β: -0.34; 95% CI -0.71, 0.02; p = 0.07 for CDR-Sb/ β: -2.00; -5.70, 1.70; p = 0.29 for DAD). Further, ongoing statin use was not associated with adverse events, serious adverse events, unscheduled GP visits, or unscheduled hospitalisation. CONCLUSION Ongoing statin use was not associated with cognitive decline or dementia progression in mild-moderate AD. Similarly, use was not associated with adverse events including abnormal liver function tests or falls. Whilst safe in those with AD, the current results suggest ongoing statin use does not delay cognitive decline or clinical progression in established AD.
Collapse
Affiliation(s)
- Claire Murphy
- Department of Age-Related Healthcare, Tallaght University Hospital, Dublin, Ireland
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Adam H Dyer
- Department of Age-Related Healthcare, Tallaght University Hospital, Dublin, Ireland
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Brian Lawlor
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Sean P Kennelly
- Department of Age-Related Healthcare, Tallaght University Hospital, Dublin, Ireland
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
32
|
Aaron SE, Tomoto T, Zhang R, Thyfault JP, Vidoni ED, Montgomery RN, Burns JM, Billinger SA. Statin contribution to middle cerebral artery blood flow velocity in older adults at risk for dementia. Eur J Appl Physiol 2022; 122:2417-2426. [PMID: 35960268 PMCID: PMC9830632 DOI: 10.1007/s00421-022-05022-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/02/2022] [Indexed: 01/12/2023]
Abstract
PURPOSE It is plausible that statins could improve cerebral blood flow through pleiotropic mechanisms. The purpose of this investigation was to assess the contribution of statins to cerebrovascular variables in older adults with dyslipidemia and familial history of dementia. Furthermore, we explored the interaction between statin use and sex due to prevalent bias in statin trials. METHODS Middle cerebral artery blood flow velocity (MCAv) was measured using transcranial Doppler ultrasound. Continuous supine rest recordings lasted 8 min. Participants included in analyses were statin (n = 100) or non-statin users (n = 112). RESULTS MCAv and cerebrovascular conductance were significantly higher in statin users (p = 0.047; p = 0.04), and pulsatility index (PI) was significantly lower in statin users (p < 0.01). An interaction effect between statin use and sex was present for PI (p = 0.02); female statin users had significantly lower cerebrovascular resistance than the other three groups. CONCLUSION In this cross-sectional analysis, statin use was positively associated with cerebrovascular variables in older adults at risk for dementia. Female statin users had significantly higher resting MCAv and cerebrovascular conductance than female non-statin users. The greatest contribution of statin use was the association with reduced cerebrovascular resistance. Given that cerebrovascular dysregulation is one of the earliest changes in Alzheimer's disease and related dementia pathology, targeting the cerebrovasculature with statins may be a promising prevention strategy.
Collapse
Affiliation(s)
- Stacey E Aaron
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tsubasa Tomoto
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, TX, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Ibaraki, Japan
| | - Rong Zhang
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, Dallas, TX, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Ibaraki, Japan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, KS, USA
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, MO, USA
- University of Kansas Alzheimer's Research Disease Center, Fairway, KS, USA
| | - Eric D Vidoni
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer's Research Disease Center, Fairway, KS, USA
| | - Robert N Montgomery
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeffrey M Burns
- University of Kansas Alzheimer's Research Disease Center, Fairway, KS, USA
| | - Sandra A Billinger
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.
- University of Kansas Alzheimer's Research Disease Center, Fairway, KS, USA.
- Department of Physical Medicine and Rehabilitation, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
33
|
Cerami C, Perini G, Panzavolta A, Cotta Ramusino M, Costa A. A Call for Drug Therapies for the Treatment of Social Behavior Disorders in Dementia: Systematic Review of Evidence and State of the Art. Int J Mol Sci 2022; 23:ijms231911550. [PMID: 36232852 PMCID: PMC9569533 DOI: 10.3390/ijms231911550] [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: 08/18/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/14/2022] Open
Abstract
Growing evidence supports the presence of social cognition deficits and social behavior alterations in major and minor neurocognitive disorders (NCDs). Even though the ability to identify socio-emotional changes has significantly improved in recent years, there is still no specific treatment available. Thus, we explored evidence of drug therapies targeting social cognition alterations in NCDs. Papers were selected according to PRISMA guidelines by searching on the PubMed and Scopus databases. Only papers reporting information on pharmacological interventions for the treatment of social cognition and/or social behavioral changes in major and/or minor NCDs were included. Among the 171 articles entered in the paper selection, only 9 papers were eligible for the scope of the review. Trials testing pharmacological treatments for socio-emotional alterations in NCDs are poor and of low-medium quality. A few attempts with neuroprotective, psychoactive, or immunomodulating drugs have been made. Oxytocin is the only drug specifically targeting the social brain that has been tested with promising results in frontotemporal dementia. Its beneficial effects in long-term use have yet to be evaluated. No recommendation can currently be provided. There is a long way to go to identify and test effective targets to treat social cognition changes in NCDs for the ultimate benefit of patients and caregivers.
Collapse
Affiliation(s)
- Chiara Cerami
- IUSS Cognitive Neuroscience (ICoN) Center, University School for Advanced Studies IUSS, 27100 Pavia, Italy
- Cognitive Computational Neuroscience Research Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy
- Correspondence:
| | - Giulia Perini
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, 27100 Pavia, Italy
- Dementia Research Center (DRC), IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Andrea Panzavolta
- IUSS Cognitive Neuroscience (ICoN) Center, University School for Advanced Studies IUSS, 27100 Pavia, Italy
| | - Matteo Cotta Ramusino
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, 27100 Pavia, Italy
- Dementia Research Center (DRC), IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Alfredo Costa
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, 27100 Pavia, Italy
- Dementia Research Center (DRC), IRCCS Mondino Foundation, 27100 Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
34
|
Statins Use in Alzheimer Disease: Bane or Boon from Frantic Search and Narrative Review. Brain Sci 2022; 12:brainsci12101290. [PMID: 36291224 PMCID: PMC9599431 DOI: 10.3390/brainsci12101290] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer’s disease (AD) was used to describe pre-senile dementia to differentiate it from senile dementia, which develops in the adult age group of more than 65 years. AD is characterized by the deposition of amyloid beta (Aβ) plaque and tau-neurofibrillary tangles (TNTs) in the brain. The neuropathological changes in AD are related to the deposition of amyloid plaques, neurofibrillary tangles, and progression of neuroinflammation, neuronal mitochondrial dysfunction, autophagy dysfunction, and cholinergic synaptic dysfunction. Statins are one of the main cornerstone drugs for the management of cardiovascular disorders regardless of dyslipidemia status. Increasing the use of statins, mainly in the elderly groups for primary and secondary prevention of cardiovascular diseases, may affect their cognitive functions. Extensive and prolonged use of statins may affect cognitive functions in healthy subjects and dementia patients. Statins-induced cognitive impairments in both patients and health providers had been reported according to the post-marketing survey. This survey depends mainly on sporadic cases, and no cognitive measures were used. Evidence from prospective and observational studies gives no robust conclusion regarding the beneficial or detrimental effects of statins on cognitive functions in AD patients. Therefore, this study is a narrative review aimed with evidences to the beneficial, detrimental, and neutral effects of statins on AD.
Collapse
|
35
|
Degrush E, Shazeeb MS, Drachman D, Vardar Z, Lindsay C, Gounis MJ, Henninger N. Cumulative effect of simvastatin, L-arginine, and tetrahydrobiopterin on cerebral blood flow and cognitive function in Alzheimer's disease. Alzheimers Res Ther 2022; 14:134. [PMID: 36115980 PMCID: PMC9482313 DOI: 10.1186/s13195-022-01076-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVES Vascular disease is a known risk factor for Alzheimer's disease (AD). Endothelial dysfunction has been linked to reduced cerebral blood flow. Endothelial nitric oxide synthase pathway (eNOS) upregulation is known to support endothelial health. This single-center, proof-of-concept study tested whether the use of three medications known to augment the eNOS pathway activity improves cognition and cerebral blood flow (CBF). METHODS Subjects with mild AD or mild cognitive impairment (MCI) were sequentially treated with the HMG-CoA reductase synthesis inhibitor simvastatin (weeks 0-16), L-arginine (weeks 4-16), and tetrahydrobiopterin (weeks 8-16). The primary outcome of interest was the change in CBF as measured by MRI from baseline to week 16. Secondary outcomes included standard assessments of cognition. RESULTS A total of 11 subjects were deemed eligible and enrolled. One subject withdrew from the study after enrollment, leaving 10 subjects for data analysis. There was a significant increase in CBF from baseline to week 8 by ~13% in the limbic and ~15% in the cerebral cortex. Secondary outcomes indicated a modest but significant increase in the MMSE from baseline (24.2±3.2) to week 16 (26.0±2.7). Exploratory analysis indicated that subjects with cognitive improvement (reduction of the ADAS-cog 13) had a significant increase in their respective limbic and cortical CBF. CONCLUSIONS Treatment of mild AD/MCI subjects with medications shown to augment the eNOS pathway was well tolerated and associated with modestly increased cerebral blood flow and cognitive improvement. TRIAL REGISTRATION This study is registered in https://www. CLINICALTRIALS gov ; registration identifier: NCT01439555; date of registration submitted to registry: 09/23/2011; date of first subject enrollment: 11/2011.
Collapse
Affiliation(s)
- Elizabeth Degrush
- Department of Neurology, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA.
- Department of Psychiatry, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA.
| | - Mohammed Salman Shazeeb
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- New England Center for Stroke Research, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - David Drachman
- Department of Neurology, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA
| | - Zeynep Vardar
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- New England Center for Stroke Research, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Clifford Lindsay
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Matthew J Gounis
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- New England Center for Stroke Research, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Nils Henninger
- Department of Neurology, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA
- Department of Psychiatry, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA
| |
Collapse
|
36
|
Rudajev V, Novotny J. Cholesterol as a key player in amyloid β-mediated toxicity in Alzheimer’s disease. Front Mol Neurosci 2022; 15:937056. [PMID: 36090253 PMCID: PMC9453481 DOI: 10.3389/fnmol.2022.937056] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder that is one of the most devastating and widespread diseases worldwide, mainly affecting the aging population. One of the key factors contributing to AD-related neurotoxicity is the production and aggregation of amyloid β (Aβ). Many studies have shown the ability of Aβ to bind to the cell membrane and disrupt its structure, leading to cell death. Because amyloid damage affects different parts of the brain differently, it seems likely that not only Aβ but also the nature of the membrane interface with which the amyloid interacts, helps determine the final neurotoxic effect. Because cholesterol is the dominant component of the plasma membrane, it plays an important role in Aβ-induced toxicity. Elevated cholesterol levels and their regulation by statins have been shown to be important factors influencing the progression of neurodegeneration. However, data from many studies have shown that cholesterol has both neuroprotective and aggravating effects in relation to the development of AD. In this review, we attempt to summarize recent findings on the role of cholesterol in Aβ toxicity mediated by membrane binding in the pathogenesis of AD and to consider it in the broader context of the lipid composition of cell membranes.
Collapse
|
37
|
Duan Y, Gong K, Xu S, Zhang F, Meng X, Han J. Regulation of cholesterol homeostasis in health and diseases: from mechanisms to targeted therapeutics. Signal Transduct Target Ther 2022; 7:265. [PMID: 35918332 PMCID: PMC9344793 DOI: 10.1038/s41392-022-01125-5] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Disturbed cholesterol homeostasis plays critical roles in the development of multiple diseases, such as cardiovascular diseases (CVD), neurodegenerative diseases and cancers, particularly the CVD in which the accumulation of lipids (mainly the cholesteryl esters) within macrophage/foam cells underneath the endothelial layer drives the formation of atherosclerotic lesions eventually. More and more studies have shown that lowering cholesterol level, especially low-density lipoprotein cholesterol level, protects cardiovascular system and prevents cardiovascular events effectively. Maintaining cholesterol homeostasis is determined by cholesterol biosynthesis, uptake, efflux, transport, storage, utilization, and/or excretion. All the processes should be precisely controlled by the multiple regulatory pathways. Based on the regulation of cholesterol homeostasis, many interventions have been developed to lower cholesterol by inhibiting cholesterol biosynthesis and uptake or enhancing cholesterol utilization and excretion. Herein, we summarize the historical review and research events, the current understandings of the molecular pathways playing key roles in regulating cholesterol homeostasis, and the cholesterol-lowering interventions in clinics or in preclinical studies as well as new cholesterol-lowering targets and their clinical advances. More importantly, we review and discuss the benefits of those interventions for the treatment of multiple diseases including atherosclerotic cardiovascular diseases, obesity, diabetes, nonalcoholic fatty liver disease, cancer, neurodegenerative diseases, osteoporosis and virus infection.
Collapse
Affiliation(s)
- Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Suowen Xu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Feng Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xianshe Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China. .,College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.
| |
Collapse
|
38
|
Magyari A, Ye M, Margolis DJ, McCulloch CE, Cummings SR, Yaffe K, Langan SM, Abuabara K. Adult atopic eczema and the risk of dementia: A population-based cohort study. J Am Acad Dermatol 2022; 87:314-322. [PMID: 35367295 DOI: 10.1016/j.jaad.2022.03.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chronic inflammatory conditions have been linked to dementia, but little is known about the role of atopic eczema, an inflammatory condition recently recognized to be common among older adults. OBJECTIVE To determine whether active atopic eczema is associated with incident dementia. METHODS A longitudinal cohort study of 1,767,667 individuals aged 60 to 99 years registered with The Health Improvement Network, a primary care cohort in the United Kingdom. The diagnoses of atopic eczema and dementia were identified using medical record codes. RESULTS The incidence of dementia was 57 per 10,000 person-years among those with atopic eczema during follow-up (12.1% of the population) compared with 44 per 10,000 person-years in the control group. This translated to a 27% increased risk of dementia (hazard ratio, 1.27; 95% CI, 1.23-1.30) in adjusted Cox proportional hazard models. Similar associations were observed in subgroup analyses of vascular dementia and Alzheimer's disease. The association persisted after additionally adjusting for the use of systemic corticosteroids (hazard ratio, 1.29; 95% CI, 1.26-1.33) and potential mediators (hazard ratio, 1.19; 95% CI, 1.16-1.22). More severe eczema was associated with a higher risk of dementia. LIMITATIONS Lack of detailed data on severity. CONCLUSION Atopic eczema was associated with a small but increased risk of incident dementia. The association increased with the severity of atopic eczema.
Collapse
Affiliation(s)
- Alexa Magyari
- UC Berkeley School of Public Health, Berkeley, California
| | - Morgan Ye
- Department of Dermatology, University of California, San Francisco School of Medicine (UCSF), San Francisco, California
| | - David J Margolis
- Department of Dermatology and Center for Epidemiology and Biostatistics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Charles E McCulloch
- Division of Biostatistics, University of California, San Francisco School of Medicine (UCSF), San Francisco, California
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Kristine Yaffe
- Center for Population Brain Health, University of California, San Francisco School of Medicine (UCSF), San Francisco, California
| | - Sinéad M Langan
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
| | - Katrina Abuabara
- UC Berkeley School of Public Health, Berkeley, California; Department of Dermatology, University of California, San Francisco School of Medicine (UCSF), San Francisco, California.
| |
Collapse
|
39
|
Abstract
Hyperlipidemia is common in patients with intracerebral hemorrhage (ICH). Accumulating evidence indicates that patients with ICH are at risk for future hemorrhage recurrence, cardiovascular disease, and ischemic stroke and highlights the importance of secondary prevention of vascular events after ICH. Although the benefits of intensive treatment of hyperlipidemia for reducing ischemic cardiac and vascular events in patients with ischemic stroke are well established, the benefit versus harm in patients with ICH are less clear. Epidemiological studies suggest that hyperlipidemia is protective against ICH and that intensive lowering of lipids is associated with increased risk for ICH. Similarly, although currently available lipid-lowering treatments have been thoroughly studied in patients with ischemic cardiac and vascular disease, only few randomized trials of these therapies included a very small number of patients with history of ICH. Thus, limiting any definitive conclusions regarding the safety and net benefit of these treatments in ICH populations. Currently, there is no consensus regarding the optimal strategy for management of hyperlipidemia after ICH. In this article, we review relevant literature to outline the competing risks and benefits of lipid-lowering treatments in this vulnerable patient population. We suggest a treatment paradigm based on available data but note that data from dedicated randomized trials are needed to build the necessary evidence to guide optimal lipid-lowering strategy in patients with a history of ICH.
Collapse
Affiliation(s)
- Ashkan Shoamanesh
- McMaster University / Population Health Research Institute, Dept. of Medicine, Hamilton, ON, CA
| | - Magdy Selim
- Beth Israel Deaconess Medical Center / Harvard Medical School, Dept. of Neurology, Stroke Division, Boston, MA
| |
Collapse
|
40
|
Jordan KL, Koss DJ, Outeiro TF, Giorgini F. Therapeutic Targeting of Rab GTPases: Relevance for Alzheimer's Disease. Biomedicines 2022; 10:1141. [PMID: 35625878 PMCID: PMC9138223 DOI: 10.3390/biomedicines10051141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/22/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022] Open
Abstract
Rab GTPases (Rabs) are small proteins that play crucial roles in vesicle transport and membrane trafficking. Owing to their widespread functions in several steps of vesicle trafficking, Rabs have been implicated in the pathogenesis of several disorders, including cancer, diabetes, and multiple neurodegenerative diseases. As treatments for neurodegenerative conditions are currently rather limited, the identification and validation of novel therapeutic targets, such as Rabs, is of great importance. This review summarises proof-of-concept studies, demonstrating that modulation of Rab GTPases in the context of Alzheimer's disease (AD) can ameliorate disease-related phenotypes, and provides an overview of the current state of the art for the pharmacological targeting of Rabs. Finally, we also discuss the barriers and challenges of therapeutically targeting these small proteins in humans, especially in the context of AD.
Collapse
Affiliation(s)
- Kate L. Jordan
- Department of Genetics and Genome Biology, University of Leicester, University Road, Leicester LE1 7RH, UK;
| | - David J. Koss
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK; (D.J.K.); (T.F.O.)
| | - Tiago F. Outeiro
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK; (D.J.K.); (T.F.O.)
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany
- Max Planck Institute for Natural Sciences, 37075 Göttingen, Germany
- Scientific Employee with a Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075 Göttingen, Germany
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, University Road, Leicester LE1 7RH, UK;
| |
Collapse
|
41
|
Bai X, Mai M, Yao K, Zhang M, Huang Y, Zhang W, Guo X, Xu Y, Zhang Y, Qurban A, Duan L, Bu J, Zhang J, Wu J, Zhao Y, Yuan X, Zu H. The role of DHCR24 in the pathogenesis of AD: re-cognition of the relationship between cholesterol and AD pathogenesis. Acta Neuropathol Commun 2022; 10:35. [PMID: 35296367 PMCID: PMC8925223 DOI: 10.1186/s40478-022-01338-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/27/2022] [Indexed: 02/01/2023] Open
Abstract
Previous studies show that 3β-hydroxysterol-Δ24 reductase (DHCR24) has a remarked decline in the brain of AD patients. In brain cholesterol synthetic metabolism, DHCR24 is known as the heavily key synthetase in cholesterol synthesis. Moreover, mutations of DHCR24 gene result in inhibition of the enzymatic activity of DHCR24, causing brain cholesterol deficiency and desmosterol accumulation. Furthermore, in vitro studies also demonstrated that DHCR24 knockdown lead to the inhibition of cholesterol synthesis, and the decrease of plasma membrane cholesterol and intracellular cholesterol level. Obviously, DHCR24 could play a crucial role in maintaining cholesterol homeostasis via the control of cholesterol synthesis. Over the past two decades, accumulating data suggests that DHCR24 activity is downregulated by major risk factors for AD, suggesting a potential link between DHCR24 downregulation and AD pathogenesis. Thus, the brain cholesterol loss seems to be induced by the major risk factors for AD, suggesting a possible causative link between brain cholesterol loss and AD. According to previous data and our study, we further found that the reduced cholesterol level in plasma membrane and intracellular compartments by the deficiency of DHCR24 activity obviously was involved in β-amyloid generation, tau hyperphosphorylation, apoptosis. Importantly, increasing evidences reveal that the brain cholesterol loss and lipid raft disorganization are obviously linked to neuropathological impairments which are associated with AD pathogenesis. Therefore, based on previous data and research on DHCR24, we suppose that the brain cholesterol deficiency/loss might be involved in the pathogenesis of AD.
Collapse
|
42
|
Patel KK, Sehgal VS, Kashfi K. Molecular targets of statins and their potential side effects: Not all the glitter is gold. Eur J Pharmacol 2022; 922:174906. [PMID: 35321818 PMCID: PMC9007885 DOI: 10.1016/j.ejphar.2022.174906] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/12/2022] [Accepted: 03/17/2022] [Indexed: 12/11/2022]
Abstract
Statins are a class of drugs widely used worldwide to manage hypercholesterolemia and the prevention of secondary heart attacks. Currently, available statins vary in terms of their pharmacokinetic and pharmacodynamic profiles. Although the primary target of statins is the inhibition of HMG-CoA reductase (HMGR), the rate-limiting enzyme in cholesterol biosynthesis, statins exhibit many pleiotropic effects downstream of the mevalonate pathway. These pleiotropic effects include the ability to reduce myocardial fibrosis, pathologic cardiac disease states, hypertension, promote bone differentiation, anti-inflammatory, and antitumor effects through multiple mechanisms. Although these pleiotropic effects of statins may be a cause for enthusiasm, there are many adverse effects that, for the most part, are unappreciated and need to be highlighted. These adverse effects include myopathy, new-onset type 2 diabetes, renal and hepatic dysfunction. Although these adverse effects may be relatively uncommon, considering the number of people worldwide who use statins daily, the actual number of people affected becomes quite large. Also, co-administration of statins with several other medications, herbal agents, and foods, which interact through common enzymatic pathways, can have untoward clinical consequences. In this review, we address these concerns.
Collapse
Affiliation(s)
- Kush K Patel
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Viren S Sehgal
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, USA.
| |
Collapse
|
43
|
Abstract
Although a relationship between traditional cardiovascular risk factors and stroke has long been recognized, these risk factors likely play a role in other aspects of brain health. Clinical stroke is only the tip of the iceberg of vascular brain injury that includes covert infarcts, white matter hyperintensities, and microbleeds. Furthermore, an individual's risk for not only stroke but poor brain health includes not only these traditional vascular risk factors but also lifestyle and genetic factors. The purpose of this narrative review is to summarize the state of the evidence on traditional and nontraditional vascular risk factors and their contributions to brain health. Additionally, we will review important modifiers that interact with these risk factors to increase, or, in some cases, reduce risk of adverse brain health outcomes, with an emphasis on genes and biomarkers associated with Alzheimer disease. Finally, we will consider the importance of social determinants of health in brain health outcomes.
Collapse
Affiliation(s)
- Rebecca F Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD (R.F.G.)
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UTHSA, San Antonio, TX (S.S.).,Department of Neurology, Boston University School of Medicine, Boston, MA (S.S.)
| |
Collapse
|
44
|
Seshadri S, Caunca MR, Rundek T. Vascular Dementia and Cognitive Impairment. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
45
|
Bhat A, Dalvi H, Jain H, Rangaraj N, Singh SB, Srivastava S. Perspective insights of repurposing the pleiotropic efficacy of statins in neurodegenerative disorders: An expository appraisal. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100012. [PMID: 34909647 PMCID: PMC8663947 DOI: 10.1016/j.crphar.2020.100012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 10/26/2022] Open
Abstract
Neurodegenerative disorders which affects a larger population pose a great clinical challenge. These disorders impact the quality of life of an individual by damaging the neurons, which are the unit cells of the brain. Clinicians are faced with the grave challenge of inhibiting the progression of these diseases as available treatment options fail to meet the clinical demand. Thus, treating the disease/disorder symptomatically is the Hobson's choice. The goal of the researchers is to introduce newer therapies in this segment and introducing a new molecule will take long years of development. Hence, drug repurposing/repositioning can be a better substitute in comparison to time consuming and expensive drug discovery and development cycle. Presently, a paradigm shift towards the re-purposing of drugs can be witnessed. Statins which have been previously approved as anti-hyperlipidemic agents are in the limelight of research for re-purposed drugs. Owing to their anti-inflammatory and antioxidant nature, statins act as neuroprotective in several brain disorders. Further they attenuate the amyloid plaques and protein aggregation which are the triggering factors in the Alzheimer's and Parkinson's respectively. In case of Huntington disease and Multiple sclerosis they help in improving the psychomotor symptoms and stimulate remyelination thus acting as neuroprotective. This article reviews the potential of statins in treating neurodegenerative disorders along with a brief discussion on the safety concerns associated with use of statins and human clinical trial data linked with re-tasking statins for neurodegenerative disorders along with the regulatory perspectives involved with the drug repositioning.
Collapse
Affiliation(s)
- Aditi Bhat
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Harshita Dalvi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Harsha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Nagarjun Rangaraj
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Shashi Bala Singh
- Department of Pharmacology and Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
46
|
Olmastroni E, Molari G, De Beni N, Colpani O, Galimberti F, Gazzotti M, Zambon A, Catapano AL, Casula M. Statin use and risk of dementia or Alzheimer's disease: a systematic review and meta-analysis of observational studies. Eur J Prev Cardiol 2021; 29:804-814. [PMID: 34871380 DOI: 10.1093/eurjpc/zwab208] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/10/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022]
Abstract
AIMS As the potential impact of statins on cognitive decline and dementia is still debated, we conducted a meta-analysis of observational studies to examine the effect of statin use on the risk of Alzheimer's disease (AD) and dementia. METHODS AND RESULTS PubMed, Cochrane, and EMBASE were searched since inception to January 2021. Inclusion criteria were: (i) cohort or case-control studies; (ii) statin users compared to non-users; and (iii) AD and/or dementia risk as outcome. Estimates from original studies were pooled using restricted maximum-likelihood random-effect model. Measure of effects were reported as odds ratio (OR) and 95% confidence intervals (CIs). In the pooled analyses, statins were associated with a decreased risk of dementia [36 studies, OR 0.80 (CI 0.75-0.86)] and of AD [21 studies, OR 0.68 (CI 0.56-0.81)]. In the stratified analysis by sex, no difference was observed in the risk reduction of dementia between men [OR 0.86 (CI 0.81-0.92)] and women [OR 0.86 (CI 0.81-0.92)]. Similar risks were observed for lipophilic and hydrophilic statins for both dementia and AD, while high-potency statins showed a 20% reduction of dementia risk compared with a 16% risk reduction associated with low-potency statins, suggesting a greater efficacy of the former, although a borderline statistical significance (P = 0.05) for the heterogeneity between estimates. CONCLUSION These results confirm the absence of a neurocognitive risk associated with statin treatment and suggest a potential favourable role of statins. Randomized clinical trials with an ad hoc design are needed to explore this potential neuroprotective effect.
Collapse
Affiliation(s)
- Elena Olmastroni
- Department of Pharmacological and Biomolecular Sciences, Epidemiology and Preventive Pharmacology Service (SEFAP), University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Giulia Molari
- IRCCS MultiMedica, Via Milanese 300, 20099 Sesto S. Giovanni (MI), Italy; and
| | - Noemi De Beni
- Department of Pharmacological and Biomolecular Sciences, Epidemiology and Preventive Pharmacology Service (SEFAP), University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Ornella Colpani
- Department of Pharmacological and Biomolecular Sciences, Epidemiology and Preventive Pharmacology Service (SEFAP), University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Federica Galimberti
- Department of Pharmacological and Biomolecular Sciences, Epidemiology and Preventive Pharmacology Service (SEFAP), University of Milan, Via Balzaretti 9, 20133 Milan, Italy.,IRCCS MultiMedica, Via Milanese 300, 20099 Sesto S. Giovanni (MI), Italy; and
| | - Marta Gazzotti
- Department of Pharmacological and Biomolecular Sciences, Epidemiology and Preventive Pharmacology Service (SEFAP), University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | - Alberto Zambon
- IRCCS MultiMedica, Via Milanese 300, 20099 Sesto S. Giovanni (MI), Italy; and.,Department of Medicine-DIMED, University of Padua, Via Giustiniani 2, 35128 Padua, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Epidemiology and Preventive Pharmacology Service (SEFAP), University of Milan, Via Balzaretti 9, 20133 Milan, Italy.,IRCCS MultiMedica, Via Milanese 300, 20099 Sesto S. Giovanni (MI), Italy; and
| | - Manuela Casula
- Department of Pharmacological and Biomolecular Sciences, Epidemiology and Preventive Pharmacology Service (SEFAP), University of Milan, Via Balzaretti 9, 20133 Milan, Italy.,IRCCS MultiMedica, Via Milanese 300, 20099 Sesto S. Giovanni (MI), Italy; and
| |
Collapse
|
47
|
Dutta S, Rahman S, Ahmad R, Kumar T, Dutta G, Banerjee S, Abubakar AR, Rowaiye AB, Dhingra S, Ravichandiran V, Kumar S, Sharma P, Haque M, Charan J. An evidence-based review of neuronal cholesterol role in dementia and statins as a pharmacotherapy in reducing risk of dementia. Expert Rev Neurother 2021; 21:1455-1472. [PMID: 34756134 DOI: 10.1080/14737175.2021.2003705] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Dementia is a progressive neurodegenerative disorder impairing memory and cognition. Alzheimer's Disease, followed by vascular dementia - the most typical form. Risk factors for vascular dementia include diabetes, cardiovascular disease, hyperlipidemia. Lipids' levels are significantly associated with vascular changes in the brain. AREAS COVERED The present article reviews the cholesterol metabolism in the brain, which includes: the synthesis, transport, storage, and elimination process. Additionally, it reviews the role of cholesterol in the pathogenesis of dementia and statin as a therapeutic intervention in dementia. In addition to the above, it further reviews evidence in support of as well as against statin therapy in dementia, recent updates of statin pharmacology, and demerits of use of statin pharmacotherapy. EXPERT OPINION Amyloid-β peptides and intraneuronal neurofibrillary tangles are markers of Alzheimer's disease. Evidence shows cholesterol modulates the functioning of enzymes associated with Amyloid-β peptide processing and synthesis. Lowering cholesterol using statin may help prevent or delay the progression of dementia. This paper reviews the role of statin in dementia and recommends extensive future studies, including genetic research, to obtain a precise medication approach for patients with dementia.
Collapse
Affiliation(s)
- Siddhartha Dutta
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujrat, India
| | - Sayeeda Rahman
- School of Medicine, American University of Integrative Sciences, Bridgetown, Barbados
| | - Rahnuma Ahmad
- Department of Physiology, Medical College for Women and Hospital, Dhaka, Bangladesh
| | - Tarun Kumar
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Jodhpur, Rajasthan, India
| | - Gitashree Dutta
- Department of Community Medicine, Neigrihms, Shillong, India
| | | | - Abdullahi Rabiu Abubakar
- Department of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Bayero University, Kano, Nigeria
| | - Adekunle Babajide Rowaiye
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka, Nigeria
| | - Sameer Dhingra
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Kolkata, Kolkata, India
| | - Santosh Kumar
- Department of Periodontology and Implantology, Karnavati University, Gandhinagar, India
| | - Paras Sharma
- Department of Pharmacognosy, BVM College of Pharmacy, Gwalior, India
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sungai Besi, Kuala Lumpur, Malaysia
| | - Jaykaran Charan
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujrat, India
| |
Collapse
|
48
|
Environmental Pyrethroid Exposure and Cognitive Dysfunction in U.S. Older Adults: The NHANES 2001-2002. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182212005. [PMID: 34831761 PMCID: PMC8623149 DOI: 10.3390/ijerph182212005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/13/2021] [Accepted: 11/13/2021] [Indexed: 11/17/2022]
Abstract
Pyrethroid compounds are widely used in household insecticides and agricultural pesticides. Recent studies, however, report that pyrethroid exposures affect neurobehavioral function in animals and may be associated with adverse neurocognitive development in children. This study aimed to examine the association between pyrethroid exposure and cognitive dysfunction in older adults using a well-defined general population. We analyzed data from 336 individuals, aged 60–84 years, who participated in the National Health and Nutrition Examination Survey 2001–2002. We used urinary 3-phenoxybenzoic acid (3-PBA) concentration as a biomarker of pyrethroid exposures and assessed cognitive function with the digit–symbol coding test. The geometric means (±geometric standard errors) of creatinine-uncorrected and corrected urinary 3-PBA were 0.30 (±0.87) μg/L and 0.36 (±0.89) μg/g. After adjusting for sociodemographic factors, higher 3-PBA concentrations (> vs. ≤0.30 μg/g creatinine (median)) were associated with lower scores of cognitive function (−3.83 95% confidence interval: −7.11, −0.54). Significance was persistent after additionally adjusting for physical activity and smoking pack-year (−3.76 95% CI: −7.16, −0.36) and further adjusting for BMI and presence of hypertension and diabetes (−3.82 95% CI: −6.92, −0.71). Our findings suggest that pyrethroid exposure is associated with cognitive dysfunction in older adults.
Collapse
|
49
|
Trzeciak P, Herbet M, Dudka J. Common Factors of Alzheimer's Disease and Rheumatoid Arthritis-Pathomechanism and Treatment. Molecules 2021; 26:6038. [PMID: 34641582 PMCID: PMC8512006 DOI: 10.3390/molecules26196038] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/20/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022] Open
Abstract
The accumulation of amyloid plaques, or misfolded fragments of proteins, leads to the development of a condition known as amyloidosis, which is clinically recognized as a systemic disease. Amyloidosis plays a special role in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease, and rheumatoid arthritis (RA). The occurrence of amyloidosis correlates with the aging process of the organism, and since nowadays, old age is determined by the comfort of functioning and the elimination of unpleasant disease symptoms in the elderly, exposure to this subject is justified. In Alzheimer's disease, amyloid plaques negatively affect glutaminergic and cholinergic transmission and loss of sympathetic protein, while in RA, amyloids stimulated by the activity of the immune system affect the degradation of the osteoarticular bond. The following monograph draws attention to the over-reactivity of the immune system in AD and RA, describes the functionality of the blood-brain barrier as an intermediary medium between RA and AD, and indicates the direction of research to date, focusing on determining the relationship and the cause-effect link between these disorders. The paper presents possible directions for the treatment of amyloidosis, with particular emphasis on innovative therapies.
Collapse
Affiliation(s)
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090 Lublin, Poland; (P.T.)
| | | |
Collapse
|
50
|
Mid- and Late-Life Migraine Is Associated with an Increased Risk of All-Cause Dementia and Alzheimer's Disease, but Not Vascular Dementia: A Nationwide Retrospective Cohort Study. J Pers Med 2021; 11:jpm11100990. [PMID: 34683131 PMCID: PMC8540823 DOI: 10.3390/jpm11100990] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/06/2023] Open
Abstract
We used a nationwide cohort sample of data from 2002 to 2013, representing approximately 1 million patients to investigate the prospective association between migraine and dementia. The migraine group (n = 1472) included patients diagnosed between 2002 and 2004, aged over 55 years; the comparison group was selected using propensity score matching (n = 5888). Cox proportional hazards regression analyses was used to calculate the hazard ratios (HRs). The incidence of dementia was 13.5 per 1000 person-years in the migraine group. Following adjustment for sociodemographic and comorbidities variables, patients with migraine developed dementia more frequently than those in the comparison group (adjusted HR = 1.37, 95% confidence interval [CI], 1.16–1.61). In the subgroup analysis, we found a higher HR of dementia events in male, the presence of comorbidities, and older age (≥65) patients with migraine, compared to those without migraine. Moreover, patients with migraine had a significantly higher incidence of Alzheimer’s disease (adjusted HR = 1.31, 95% CI, 1.08–1.58), but not vascular dementia, than those without migraine. Therefore, our findings suggest that mid- and late-life migraines may be associated with an increased incidence of all-cause dementia and Alzheimer’s disease, but not vascular dementia.
Collapse
|