1
|
Prajapati SK, Wang S, Mishra SP, Jain S, Yadav H. Protection of Alzheimer's disease progression by a human-origin probiotics cocktail. Sci Rep 2025; 15:1589. [PMID: 39794404 PMCID: PMC11724051 DOI: 10.1038/s41598-024-84780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Microbiome abnormalities (dysbiosis) significantly contribute to the progression of Alzheimer's disease (AD). However, the therapeutic efficacy of microbiome modulators in protecting against these ailments remains poorly studied. Herein, we tested a cocktail of unique probiotics, including 5 Lactobacillus and 5 Enterococcus strains isolated from infant gut with proven microbiome modulating capabilities. We aimed to determine the probiotics cocktail's efficacy in ameliorating AD pathology in a humanized AD mouse model of APP/PS1 strains. Remarkably, feeding mice with 1 × 1011 CFU per day in drinking water for 16 weeks significantly reduced cognitive decline (measured by the Morris Water Maze test) and AD pathology markers, such as Aβ aggregation, microglia activation, neuroinflammation, and preserved blood-brain barrier (BBB) tight junctions. The beneficial effects were linked to a reduced inflammatory microbiome, leading to decreased gut permeability and inflammation in both systemic circulation and the brain. Although both male and female mice showed overall improvements in cognition and biological markers, females did not exhibit improvements in specific markers related to inflammation and barrier permeability, suggesting that the underlying mechanisms may differ depending on sex. In conclusion, our results suggest that this unique probiotics cocktail could serve as a prophylactic agent to reduce the progression of cognitive decline and AD pathology. This is achieved by beneficially modulating the microbiome, improving intestinal tight junction proteins, reducing permeability in both gut and BBB, and decreasing inflammation in the gut, blood circulation, and brain, ultimately mitigating AD pathology and cognitive decline.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shaohua Wang
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Biomedical Sciences, Infectious and Tropical Disease Institute, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Sidharth P Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Department of Internal Medicine-Digestive Diseases and Nutrition, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
2
|
Hori K, Yoshimura Y, Wakabayashi H, Nagano F, Matsumoto A, Shimazu S, Shiraishi A, Kido Y, Bise T, Kuzuhara A, Hamada T, Yoneda K, Maekawa K. Improved Systemic Inflammation is Associated with Functional Prognosis in Post-Stroke Patients. Ann Geriatr Med Res 2024; 28:388-394. [PMID: 38724451 PMCID: PMC11695764 DOI: 10.4235/agmr.24.0020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/16/2024] [Accepted: 04/26/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Systemic inflammation is associated with poor functional outcomes. However, the effects of improved inflammation on functional indicators remain unclear. This study aimed to clarify the relationship between improvements in systemic inflammation and activities of daily living in patients after stroke. METHODS This retrospective cohort study included patients post stroke with systemic inflammation upon admission. Systemic inflammation was defined as a modified Glasgow Prognostic Score (mGPS) score of 1-2. Improvement in systemic inflammation was defined as a reduction in mGPS score or blood C-reactive protein (CRP) levels during hospitalization. The primary outcomes were the motor items of the Functional Independence Measure (FIM-motor) at discharge. We applied multiple linear regression analysis to examine whether reduced systemic inflammation was associated with outcomes after adjusting for confounding factors. RESULTS Of the 1,490 patients recruited, 158 (median age of 79 years; 88 men) had systemic inflammation on admission and were included in the study. Among these patients, 131 (82.9%) and 147 (93.0%) exhibited reduced mGPS and CRP levels, respectively. The median change in CRP was 2.1 mg/dL (interquartile range, 1.1-3.8). Multivariate analysis revealed that improvements in mGPS (β=0.125, p=0.012) and CRP levels (β=0.108, p=0.108) were independently and positively associated with FIM-motor at discharge. CONCLUSIONS Improvement in systemic inflammation was positively associated with functional outcomes in patients post stroke. Early detection and therapeutic intervention for systemic inflammation may further improve outcomes in these patients.
Collapse
Affiliation(s)
- Kota Hori
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Yoshihiro Yoshimura
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Hidetaka Wakabayashi
- Department of Rehabilitation Medicine, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Fumihiko Nagano
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Ayaka Matsumoto
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Sayuri Shimazu
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Ai Shiraishi
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Yoshifumi Kido
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Takahiro Bise
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Aomi Kuzuhara
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Takenori Hamada
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Kouki Yoneda
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| | - Kenichiro Maekawa
- Center for Sarcopenia Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto, Japan
| |
Collapse
|
3
|
van Lent DM, Jacques PF, Charisis SM, Mesa HG, Satizabal C, Yuan C, Vasan RS, Seshadri S, Beiser A, Himali JJ, Jacob ME. Dietary inflammatory index scores and cognitive aging: results from the Framingham heart Study offspring cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.27.24318084. [PMID: 39649578 PMCID: PMC11623757 DOI: 10.1101/2024.11.27.24318084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Background Nutritional factors can abet or protect against systemic chronic inflammation, which plays an important role in the development and progression of dementia. We evaluated whether higher (i.e. pro-inflammatory) Dietary Inflammatory Index (DII) scores were associated with cognitive decline in the community-based Offspring Framingham Heart Study (FHS). Method 1,614 older adults (mean age 61 years [standard deviation (SD)], 9;55% women]) completed validated 126-item Food Frequency Questionnaires (FFQ), administered at FHS examination cycle 7 (1998-2001) and examination cycle 5 (1991-1995) and/or 6 (1995-1998). We created a DII score (based on the published method by Shivappa et al. 2014) for each available exam; a cumulative DII score was calculated by averaging exam-specific scores across the two or three exams. Cognitive testing was completed at call back sessions following examination cycles 7 and 8 (2005-2008). Exam 7 was considered as study baseline and participants were followed over a mean time of seven years (SD 1). We excluded participants with prevalent dementia at baseline and those with no cognitive testing data at either/or exams 7 and 8. We examined associations between the cumulative DII score and cognitive test scores over time using annualized change adjusting for age, sex, education (model 1) and additionally for exam 7 measures of body mass index, total energy intake, total cholesterol: high-density lipoprotein ratio, smoking and anti-cholesterol medication (model 2). Results Higher DII scores were significantly associated with (less) decline in performance on Similarities (verbal comprehension/reasoning) and Global cognition, following adjustments for model 1 covariates (Model 1:β and SE, 0.017, 0.008,p=0.03; 0.004,0.002,p=0.02, respectively). The effect sizes remained similar after additional adjustment for Model 2 covariates (0.018, 0.009,p=0.06; 0.004, 0.002,p=0.03, respectively). Additionally, we found that higher DII scores associated with accelerated decline in performance on Trail Making B-A (processing speed and executive function) (Model 2: -0.010, 0.004,p=0.03). We observed no relationships between higher DII scores and other neuropsychological tests. Further, stratified analyses revealed a linear relationship between higher DII scores and (less) decline in performance on Hooper visual organization among men, but not women (Model 2: 0.022, 0.010,p=0.02; -0.011, 0.009,p=0.23, respectively). Conclusion Higher DII scores were associated with (less) cognitive decline. We take our unexpected findings with caution as we previous have seen a relationship between higher DII scores and increased risk for dementia. To date, such studies have been very limited, most studies that found a relationship were cross-sectional and have used less sensitive testing. Future longitudinal studies with sensitive neuropsychological test measures are encouraged to elucidate whether a longitudinal relationship between higher DII scores and age-related cognitive decline exists.
Collapse
|
4
|
Kadam R, Gupta M, Lazarov O, Prabhakar BS. Brain-immune interactions: implication for cognitive impairments in Alzheimer's disease and autoimmune disorders. J Leukoc Biol 2024; 116:1269-1290. [PMID: 38869088 DOI: 10.1093/jleuko/qiae134] [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: 11/13/2023] [Revised: 05/09/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024] Open
Abstract
Progressive memory loss and cognitive dysfunction, encompassing deficits in learning, memory, problem solving, spatial reasoning, and verbal expression, are characteristics of Alzheimer's disease and related dementia. A wealth of studies has described multiple roles of the immune system in the development or exacerbation of dementia. Individuals with autoimmune disorders can also develop cognitive dysfunction, a phenomenon termed "autoimmune dementia." Together, these findings underscore the pivotal role of the neuroimmune axis in both Alzheimer's disease and related dementia and autoimmune dementia. The dynamic interplay between adaptive and innate immunity, both in and outside the brain, significantly affects the etiology and progression of these conditions. Multidisciplinary research shows that cognitive dysfunction arises from a bidirectional relationship between the nervous and immune systems, though the specific mechanisms that drive cognitive impairments are not fully understood. Intriguingly, this reciprocal regulation occurs at multiple levels, where neuronal signals can modulate immune responses, and immune system-related processes can influence neuronal viability and function. In this review, we consider the implications of autoimmune responses in various autoimmune disorders and Alzheimer's disease and explore their effects on brain function. We also discuss the diverse cellular and molecular crosstalk between the brain and the immune system, as they may shed light on potential triggers of peripheral inflammation, their effect on the integrity of the blood-brain barrier, and brain function. Additionally, we assess challenges and possibilities associated with developing immune-based therapies for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Rashmi Kadam
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| |
Collapse
|
5
|
Shiraishi A, Yoshimura Y, Nagano F, Matsumoto A, Shimazu S, Kido Y, Bise T, Kuzuhara A, Hori K, Hamada T, Yoneda K, Maekawa K. Impaired oral health associations with cognitive and motor decline in activities of daily living independence during hospitalization: insights from a post-stroke cohort. J Stroke Cerebrovasc Dis 2024; 33:107966. [PMID: 39187215 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107966] [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: 02/11/2024] [Revised: 08/11/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
PURPOSE Evidence is scarce on the associations between impaired oral health and cognitive level related to independence in activities of daily living (ADLs) among hospitalized older patients. We aimed to evaluate the associations between baseline oral problems and changes in cognitive level in post-stroke patients. METHODS A retrospective cohort study was conducted, encompassing consecutively hospitalized post-stroke patients. Revised Oral Assessment Guide (ROAG) as a measure of oral health and function was assessed at admission. Primary outcomes included discharge scores for cognitive function assessed by the cognitive domain of the Functional Independence Measure (FIM-cognition) and the corresponding change in FIM-cognition during hospitalization. Another outcome measure was the motor domain of FIM (FIM-motor). Multivariate linear regression analyses were employed to assess the association between baseline ROAG and the designated outcomes, adjusting for potential confounding factors. RESULTS Data from 955 patients (mean age 73.2 years; 53.6% men) were included in the analysis. The median ROAG was 10 [9, 12], with 811 patients (84.9%) presenting oral problems. After fully adjusting for confounding factors, the ROAG was significantly and negatively associated with FIM-cognition at discharge (β = -0.107, p = 0.031) and FIM-cognition gain (β = -0.093, p = 0.018). Further, the ROAG was independently and negatively associated with FIM-motor at discharge (β = -0.043, p = 0.013) and FIM-motor gain (β = -0.065, p = 0.013). CONCLUSION Oral problems were associated with compromised cognitive levels and a decline in physical function during the hospitalization in post-stroke patients. These results underscore the critical importance of addressing oral health in this patient population.
Collapse
Affiliation(s)
- Ai Shiraishi
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Yoshihiro Yoshimura
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Fumihiko Nagano
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Ayaka Matsumoto
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Sayuri Shimazu
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Yoshifumi Kido
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Takahiro Bise
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Aomi Kuzuhara
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Kota Hori
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Takenori Hamada
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Kouki Yoneda
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Kenichiro Maekawa
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| |
Collapse
|
6
|
Morrison L, Dyer AH, Dolphin H, Batten I, Reddy C, Widdowson M, Woods CP, Gibney J, Bourke NM, Kennelly SP. Circulating Interleukin-17A is associated with executive function in middle aged adults with and without type 2 diabetes. Brain Behav Immun Health 2024; 41:100862. [PMID: 39350951 PMCID: PMC11440310 DOI: 10.1016/j.bbih.2024.100862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/19/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
Midlife cardiovascular risk factors such as Type 2 Diabetes (T2DM) and obesity are associated with the later development of cognitive impairment and dementia. Systemic inflammation is postulated as a crucial mechanism, yet there are few studies examining this at the earliest stages prior to overt cognitive impairment. To assess this, we recruited a cohort of middle-aged cognitively-unimpaired individuals with and without uncomplicated T2DM. Comprehensive neuropsychological assessment was performed at baseline and at 4-year follow-up. Ten serum chemokines and cytokines (Eotaxin, MCP-1, MIP-1β, CXCL10, IL-6, IL-10, IL12p70, IL-17A, IFN-γ and TNF-α) were measured at both baseline and follow-up using high-sensitivity assays. Overall, 136 participants were recruited including 90 with uncomplicated midlife T2DM (age 52.6 ± 8.3; 47% female) and 46 without (age 52.9 ± 8.03; 61% female). Cognitive trajectories were stable over time and did not differ with T2DM. Yet on cross-sectional analyses at both baseline and follow-up, greater circulating IL-17A was consistently associated with poorer performance on tests of executive function/attention (β: 0.21; -0.40, -0.02, p = 0.03 at baseline; β: 0.26; -0.46, -0.05, p = 0.02 at follow-up). Associations persisted on covariate adjustment and did not differ by T2DM status. In summary, we provide evidence that greater circulating IL-17A levels were associated with poorer executive function in midlife, independent of T2DM. Long-term follow-up of this and other cohorts will further elucidate the earliest stages in the relationship between systemic inflammation and cognitive decline to provide further mechanistic insights and potentially identify those at greatest risk for later cognitive decline.
Collapse
Affiliation(s)
- Laura Morrison
- Tallaght Institute for Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Adam H Dyer
- Tallaght Institute for Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Helena Dolphin
- Tallaght Institute for Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Isabella Batten
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Conor Reddy
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
| | - Matthew Widdowson
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Conor P Woods
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - James Gibney
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
- Robert Graves Institute of Endocrinology, Tallaght University Hospital, Dublin, Ireland
| | - Nollaig M Bourke
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| | - Sean P Kennelly
- Tallaght Institute for Memory and Cognition, Tallaght University Hospital, Dublin, Ireland
- Discipline of Medical Gerontology, School of Medicine, Trinity College Dublin, Ireland
| |
Collapse
|
7
|
Geloso MC, Zupo L, Corvino V. Crosstalk between peripheral inflammation and brain: Focus on the responses of microglia and astrocytes to peripheral challenge. Neurochem Int 2024; 180:105872. [PMID: 39362496 DOI: 10.1016/j.neuint.2024.105872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
A growing body of evidence supports the link between peripheral inflammation and impairment of neurologic functions, including mood and cognitive abilities. The pathogenic event connecting peripheral inflammation and brain dysfunction is represented by neuroinflammation, a pathogenic phenomenon that provides an important contribution to neurodegeneration and cognitive decline also in Alzheimer's, Parkinson's, Huntington's diseases, as well as in Multiple Sclerosis. It is driven by resident brain immune cells, microglia and astrocytes, that acquire an activated phenotype in response to proinflammatory molecules moving from the periphery to the brain parenchyma. Although a huge progress has been made in clarifying cellular and molecular mechanisms bridging peripheral and central inflammation, a clear picture has not been achieved so far. Therefore, experimental models are of crucial relevance to clarify knowledge gaps in this regard. Many findings demonstrate that systemic inflammation induced by pathogen-associated molecular patterns, such as lipopolysaccharide (LPS), is able to trigger neuroinflammation. Therefore, LPS-administration is widely considered a useful tool to study this phenomenon. On this basis, the present review will focus on in vivo studies based on acute and subacute effects of systemic administration of LPS, with special attention on the state of art of microglia and astrocyte response to peripheral challenge.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Gemelli Science and Technology Park (GSTeP)-Organoids Research Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy.
| | - Luca Zupo
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Valentina Corvino
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| |
Collapse
|
8
|
Zhang H, Liang J, Huang J, Wang M, Wu L, Wu T, Chen N. Exerkine irisin mitigates cognitive impairment by suppressing gut-brain axis-mediated inflammation. J Adv Res 2024:S2090-1232(24)00485-5. [PMID: 39481644 DOI: 10.1016/j.jare.2024.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/16/2024] [Accepted: 10/26/2024] [Indexed: 11/02/2024] Open
Abstract
INTRODUCTION Exercise has been recognized to improve cognitive performance by optimizing gut flora and up-regulating exerkine irisin. OBJECTIVE Although exercise-induced irisin is beneficial to cognitive improvement, whether this benefit is achieved by optimizing gut microbiota and metabolites is not fully explored. METHODS After aerobic exercise and exogenous irisin interventions for 12 weeks, the 16S rRNA and metabolites in feces of 21-month-old mice were analyzed. Meanwhile, the differential miRNAs and mRNAs in hippocampal tissues were screened by high-throughput sequencing. Relevant mRNAs and proteins were evaluated by RT-PCR, Western blot, and immunofluorescence. RESULTS Compared with the young control mice, irisin levels and cognitive capacity of aged mice revealed a significant reduction, while aerobic exercise and intraperitoneal injection of exogenous irisin reversed aging-induced cognitive impairment. Similarly, 147 up-regulated and 173 down-regulated metabolites were detected in aged mice, while 64 and 45 up-regulated and 225 and 187 down-regulated metabolites were detected in aged mice with exercise and irisin interventions, respectively. Moreover, during hippocampal miRNA and mRNA sequencing analysis, 9 differential gut flora and 35 differential genes were identified to be correlated with the inflammatory signaling mediated by the TLR4/MyD88 signal pathway. CONCLUSION Aging-induced cognitive impairment is due to insulin resistance induced by TLR4/MyD88 signaling activation in hippocampal tissues mediated by gut microbiota and metabolite changes. Myokine irisin may be an important mediator in optimizing gut microbiota and metabolism for an improved understanding of mitigated aging process upon exercise interventions.
Collapse
Affiliation(s)
- Hu Zhang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jiling Liang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Jielun Huang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Minghui Wang
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Liangwen Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| | - Tong Wu
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
9
|
Shen W, Wu J, Shi L, Feng H, Yang X, Zhang Y. Explore the mechanisms of triterpenoids from Ganoderma lucidum in the protection against Alzheimer's disease via microbiota-gut-brain axis with the aid of network pharmacology. Fitoterapia 2024; 178:106150. [PMID: 39089595 DOI: 10.1016/j.fitote.2024.106150] [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: 05/02/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Ganoderma lucidum (Curtis) P. Karst.(G. lucidum) is a kind of fungi, which also a traditional Chinese medicine used for "wisdom growth" in China. Triterpenoids from G. lucidum (GLTs) are one of the main active ingredients. Based on the strategy of early intervention on Alzheimer's disease (AD) and the inextricable association between disordered gut microbiota and metabolites with AD, this study aimed to explore the mechanisms of GLTs in the protection against AD via microbiota-gut-brain axis with the aid of network pharmacology. In this study, LC-MS/MS was used to identify the main active ingredients of GLTs. Network pharmacology was used to predict the potential target and validated with Caco-2 cell model. D-galactose was used to induce the slow-onset AD on rats. Metabolomics methods basing on GC-MS combined with 16S rRNA sequencing technology was used to carry out microbiota-gut-metabolomics analysis in order to reveal the potential mechanisms of GLTs in the protection of AD. As results, GLTs showed a protection against AD effect on rats by intervening administration. The mechanisms were inextricably linked to GLTs interference with the balance of gut microbiota and metabolites. The main fecal metabolites involved were short-chain fatty acids and aromatic amino acid metabolites.
Collapse
Affiliation(s)
- Wanping Shen
- College of Medicine, Jiaxing University, Jiaxing 314001, China; Graduate school, Jilin Institute of Chemical Technology, Jilin 132022, China
| | - Jiming Wu
- College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Liyan Shi
- College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Haisong Feng
- College of Medicine, Jiaxing University, Jiaxing 314001, China
| | - Xiudong Yang
- School of Chemical and Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, China.
| | - Yan Zhang
- College of Medicine, Jiaxing University, Jiaxing 314001, China.
| |
Collapse
|
10
|
Victor-Sami S, Kamali-Roosta A, Shamsaldeen YA. Methylglyoxal induces death in human brain neuronal cells (SH-SY5Y), prevented by metformin and dapagliflozin. J Diabetes Complications 2024; 38:108832. [PMID: 39116474 DOI: 10.1016/j.jdiacomp.2024.108832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/22/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Diabetes mellitus is a metabolic disorder caused by a dysfunction in insulin action or secretion, leading to an elevation in blood glucose levels. It is a highly prevalent condition and as a result, the NHS spends 10 % of its entire budget on diabetes mellitus care, that is equivalent to £10 billion a year. Diabetes mellitus has been linked with vascular and neurological complications which may be associated with the progression of neurodegeneration and Alzheimer's disease. Chronic hyperglycaemia increases the production of the reactive oxidant species (ROS) such as methylglyoxal (MGO). MGO has been linked with vascular complications, neuropathy and cytotoxicity. The main aim of this study was to investigate the potential beneficial effect of antidiabetic agents such as metformin and dapagliflozin on human brain neuronal cells (SH-SY5Y) treated with MGO. SH-SY5Y cells were cultured in DMEM/F12 media and subjected overnight incubation with one of the following treatment conditions: Control (untreated); MGO (1 μM); MGO (100 μM); metformin (100 μM) + MGO (100 μM); and dapagliflozin (10 μM) + MGO (100 μM). Several assays were conducted to explore the effect of the treatment groups on the SH-SY5Y cells. These included: MTT assay; LDH assay, peroxynitrite fluorescence assay, and laser scanning confocal microscopy. MGO (100 μM) led to significant cell injury and damage and significantly reduced the survival of the cells by approximately 50-75 %, associated with significant increase in peroxynitrite. The addition of metformin (100 μM) or dapagliflozin (10 μM) represented significant protective effects on the cells and prevented the cell damage caused by the high MGO concentration. As a result, the findings of this research reveal that MGO-induced cell damage may partly be mediated by the generation of peroxynitrite, while the antidiabetic agents such as metformin and dapagliflozin prevent brain cell death, which potentially may play prophylactic roles against the risk of dementia in diabetic patients.
Collapse
Affiliation(s)
- Samantha Victor-Sami
- Department of applied science, school of pharmacy, University of Brighton, BN24GJ, UK
| | - Ali Kamali-Roosta
- Department of applied science, school of pharmacy, University of Brighton, BN24GJ, UK
| | - Yousif A Shamsaldeen
- Department of applied science, school of pharmacy, University of Brighton, BN24GJ, UK.
| |
Collapse
|
11
|
Duggan MR, Peng Z, Sipilä PN, Lindbohm JV, Chen J, Lu Y, Davatzikos C, Erus G, Hohman TJ, Andrews SJ, Candia J, Tanaka T, Joynes CM, Alvarado CX, Nalls MA, Cordon J, Daya GN, An Y, Lewis A, Moghekar A, Palta P, Coresh J, Ferrucci L, Kivimäki M, Walker KA. Proteomics identifies potential immunological drivers of postinfection brain atrophy and cognitive decline. NATURE AGING 2024; 4:1263-1278. [PMID: 39143319 PMCID: PMC11408246 DOI: 10.1038/s43587-024-00682-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Infections have been associated with the incidence of Alzheimer disease and related dementias, but the mechanisms responsible for these associations remain unclear. Using a multicohort approach, we found that influenza, viral, respiratory, and skin and subcutaneous infections were associated with increased long-term dementia risk. These infections were also associated with region-specific brain volume loss, most commonly in the temporal lobe. We identified 260 out of 942 immunologically relevant proteins in plasma that were differentially expressed in individuals with an infection history. Of the infection-related proteins, 35 predicted volumetric changes in brain regions vulnerable to infection-specific atrophy. Several of these proteins, including PIK3CG, PACSIN2, and PRKCB, were related to cognitive decline and plasma biomarkers of dementia (Aβ42/40, GFAP, NfL, pTau-181). Genetic variants that influenced expression of immunologically relevant infection-related proteins, including ITGB6 and TLR5, predicted brain volume loss. Our findings support the role of infections in dementia risk and identify molecular mediators by which infections may contribute to neurodegeneration.
Collapse
Affiliation(s)
- Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pyry N Sipilä
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Finnish Institute of Occupational Health, Helsinki, Finland
| | - Joni V Lindbohm
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Broad Institute of the MIT and Harvard University, The Klarman Cell Observatory, Cambridge, MA, USA
- Brain Sciences, University College London, London, UK
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yifei Lu
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Guray Erus
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shea J Andrews
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Julián Candia
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Cassandra M Joynes
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Chelsea X Alvarado
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jenifer Cordon
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Gulzar N Daya
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Alexandria Lewis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Priya Palta
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mika Kivimäki
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Brain Sciences, University College London, London, UK
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
12
|
Yoshimura Y, Nagano F, Matsumoto A, Shimazu S, Shiraishi A, Kido Y, Bise T, Kuzuhara A, Hori K, Hamada T, Yoneda K, Maekawa K. Hemoglobin levels and cognitive trajectory: unveiling prognostic insights in post-stroke geriatric cohort. J Stroke Cerebrovasc Dis 2024; 33:107856. [PMID: 38997051 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107856] [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: 02/01/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/14/2024] Open
Abstract
PURPOSE Evidence is scarce regarding the association between anemia and alterations in cognitive level among hospitalized older patients. We aimed to evaluate the associations between baseline hemoglobin (Hb) levels and changes in cognitive level in patients undergoing rehabilitation after stroke. METHODS A retrospective cohort study was conducted, encompassing consecutively hospitalized post-stroke patients. Data on serum Hb levels were extracted from medical records, specifically tests conducted within 24 hours of admission. Primary outcomes included discharge scores for cognitive function assessed by the cognitive domain of the Functional Independence Measure (FIM-cognition) and the corresponding change in FIM-cognition during hospitalization. Another outcome measure was the length of hospital stay. Multivariate linear regression analyses were employed to assess the association between Hb levels at admission and the designated outcomes, adjusting for potential confounding factors. RESULTS Data from 955 patients (mean age 73.2 years; 53.6% men) were included in the analysis. The median Hb level at admission was 13.3 [11.9, 14.5] g/dL. After fully adjusting for confounding factors, the baseline Hb level was significantly and positively associated with FIM-cognition at discharge (β = 0.045, p = 0.025) and its gain (β = 0.073, p = 0.025). Further, the baseline Hb level was independently and negatively associated with length of hospital stay (β = -0.013, p = 0.026). CONCLUSION Elevated baseline Hb levels are correlated with preserved cognitive level and shorter hospital stays in post-stroke patients. Evaluating anemia at the outset serves as a crucial prognostic indicator.
Collapse
Affiliation(s)
- Yoshihiro Yoshimura
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Fumihiko Nagano
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan
| | - Ayaka Matsumoto
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan
| | - Sayuri Shimazu
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Ai Shiraishi
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan
| | - Yoshifumi Kido
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan
| | - Takahiro Bise
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan
| | - Aomi Kuzuhara
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan
| | - Kota Hori
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan
| | - Takenori Hamada
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan.
| | - Kouki Yoneda
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan
| | - Kenichiro Maekawa
- Center for Sarcopenia and Malnutrition Research, Kumamoto Rehabilitation Hospital, Kumamoto 869-1106, Japan
| |
Collapse
|
13
|
Wang Q, Liu Y, Xu S, Liu F, Huang L, Xu F, Liu Y. Development and validation of the eMCI-CHD tool: A multivariable prediction model for the risk of mild cognitive impairment in patients with coronary heart disease. J Evid Based Med 2024; 17:535-549. [PMID: 39107928 DOI: 10.1111/jebm.12632] [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: 03/24/2024] [Accepted: 07/23/2024] [Indexed: 09/30/2024]
Abstract
OBJECTIVE This study aimed to develop and validate an eMCI-CHD tool based on clinical data to predict mild cognitive impairment (MCI) risk in patients with coronary heart disease (CHD). METHODS This cross-sectional study prospectively collected data from 400 patients with coronary heart disease (aged 55-90 years, 62% men) from July 2022 to September 2023 and randomized (7:3 ratio) them into training and validation sets. After determining the modeling variables through least absolute shrinkage and selection operator regression analysis, four ML classifiers were developed: logistic regression, extreme gradient boosting (XGBoost), support vector machine, and random forest. The performance of the models was evaluated using area under the ROC curve, accuracy, sensitivity, specificity, and F1 score. Decision curve analysis was used to assess the clinical performance of the established models. The SHapley Additive exPlanations (SHAP) method was applied to determine the significance of the features, the predictive model was visualized with a nomogram, and an online web-based calculator for predicting CHD-MCI risk scores was developed. RESULTS Of 400 CHD patients (average age 70.86 ± 8.74 years), 220 (55%) had MCI. The XGBoost model demonstrated superior performance (AUC: 0.86, accuracy: 78.57%, sensitivity: 0.74, specificity: 0.84, F1: 0.79) and underwent validation. An online tool (https://mr.cscps.com.cn/mci/index.html) with seven predictive variables (APOE gene typing, age, education, TyG index, NT-proBNP, C-reactive protein, and occupation) assessed MCI risk in CHD patients. CONCLUSION This study highlights the potential for predicting MCI risk among CHD patients using an ML model-driven nomogram and risk scoring tool based on clinical data.
Collapse
Affiliation(s)
- Qing Wang
- The Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanfei Liu
- The Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Xu
- The Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fenglan Liu
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Luqi Huang
- China Evidence-Based Medicine Center of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- The Second Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yue Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Key Laboratory of Disease and Syndrome Integration Prevention and Treatment of Vascular Aging, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
14
|
Doroszkiewicz J, Mroczko J, Winkel I, Mroczko B. Metabolic and Immune System Dysregulation: Unraveling the Connections between Alzheimer's Disease, Diabetes, Inflammatory Bowel Diseases, and Rheumatoid Arthritis. J Clin Med 2024; 13:5057. [PMID: 39274269 PMCID: PMC11396443 DOI: 10.3390/jcm13175057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/16/2024] Open
Abstract
Alzheimer's disease (AD), diabetes mellitus (DM), inflammatory bowel diseases (IBD), and rheumatoid arthritis (RA) are chronic conditions affecting millions globally. Despite differing clinical symptoms, these diseases share pathophysiological mechanisms involving metabolic and immune system dysregulation. This paper examines the intricate connections between these disorders, focusing on shared pathways such as insulin resistance, lipid metabolism dysregulation, oxidative stress, and chronic inflammation. An important aspect is the role of amyloid-beta plaques and tau protein tangles, which are hallmark features of AD. These protein aggregates are influenced by metabolic dysfunction and inflammatory processes similar to those seen in DM, RA, and IBD. This manuscript explores how amyloid and tau pathologies may be exacerbated by shared metabolic and immune dysfunction. Additionally, this work discusses the gut-brain axis and the influence of gut microbiota in mediating disease interactions. Understanding these commonalities opens new avenues for multi-targeted therapeutic approaches that address the root causes rather than merely the symptoms of these conditions. This integrative perspective could lead to more effective interventions and improved patient outcomes, emphasizing the importance of a unified approach in managing these interconnected diseases.
Collapse
Affiliation(s)
- Julia Doroszkiewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Jan Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Izabela Winkel
- Dementia Disorders Centre, Medical University of Wroclaw, 50-425 Scinawa, Poland
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
15
|
Chen X, Cai L, Fan W, Yang Q, Mao X, Yao L. Causal relationships between rheumatoid arthritis and neurodegenerative diseases: a two-sample univariable and multivariable Mendelian randomization study. Front Med (Lausanne) 2024; 11:1439344. [PMID: 39193017 PMCID: PMC11347450 DOI: 10.3389/fmed.2024.1439344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Background Observational research has highlighted a potential relationship between rheumatoid arthritis (RA) and neurodegenerative diseases (NDs). However, the confirmation of a causal connection is impeded by the inherent limitations of such studies, including vulnerability to confounding factors and the possibility of reverse causality. This study employs a two-sample Mendelian randomization (MR) approach to assess the causal impact of RA on three NDs, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Methods We aggregated data from genome-wide association studies (GWASs) targeting RA or NDs within populations of European descent. Single nucleotide polymorphisms (SNPs) with robust associations to RA were identified as instrumental variables (IVs). To estimate the association between RA and AD, PD, and ALS, we utilized the inverse variance weighted (IVW) method in our univariable MR (UVMR) analysis. Validation of the IVW results ensued through supplementary analyses using MR-Egger and weighted median methods. The multivariable MR (MVMR) analysis was conducted, adjusting for body mass index (BMI), alcohol drinking, and type 2 diabetes mellitus (T2DM). Results The UVMR analysis, based on the IVW method, revealed a significantly positive causal association between RA and late-onset (LO) AD (OR [95% CI] = 1.084 [1.020-1.153]; p = 9.980 × 10-3), while suggesting a possible inverse relationship with PD (OR [95% CI] = 0.727 [0.563-0.938]; p = 0.014). Our study did not detect any causal connections between RA and early-onset (EO) AD, atypical or mixed (AM) AD, and ALS (all p > 0.05). The MVMR analysis results indicated that after adjusting for alcohol drinking, RA remains a risk factor for LOAD (OR [95% CI] = 1.094 [1.024-1.169]; p = 0.008). However, MVMR analysis revealed no causal connections between RA and PD after adjustments for BMI, alcohol drinking, or T2DM (all p > 0.05). Sensitivity analyses showed no evidence of heterogeneity and horizontal pleiotropy. Conclusions This research provides genetic evidence indicating that RA potentially causes an increased risk of developing LOAD and PD. Such a revelation underscores the importance for individuals suffering from RA to be vigilant about the potential emergence of LOAD and PD. Ongoing monitoring and prompt detection are essential for successfully managing and intervening in this possible risk.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Neurology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Li Cai
- Department of Neurology, The Third Hospital of Changsha, Changsha, China
| | - Weibing Fan
- Department of Neurology, The Third Hospital of Changsha, Changsha, China
| | - Qian Yang
- Department of Neurology, The Third Hospital of Changsha, Changsha, China
| | - Xinfa Mao
- Department of Neurology, The Third Hospital of Changsha, Changsha, China
| | - Liping Yao
- Department of Neurology, The Third Hospital of Changsha, Changsha, China
| |
Collapse
|
16
|
Ni Y, Wang Z, Zhuge F, Zhou K, Zheng L, Hu X, Wang S, Fu O, Fu Z. Hydrolyzed Chicken Meat Extract and Its Bioactive Cyclopeptides Protect Neural Function by Attenuating Inflammation and Apoptosis via PI3K/AKT and AMPK Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:16708-16725. [PMID: 39016108 DOI: 10.1021/acs.jafc.4c02063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Cognitive decline is inevitable with age, and due to the lack of well-established pharmacotherapies for neurodegenerative disorders, dietary supplements have become important alternatives to ameliorate brain deterioration. Hydrolyzed chicken meat extract (HCE) and its bioactive components were previously found to improve neuroinflammation and cognitive decline by regulating microglia polarization. However, the effects and mechanisms of these bioactives on neurons remain unclear. Here, the most potent bioactive component on neural function in HCE was screened out, and the detailed mechanism was clarified through in vivo and in vitro experiments. We found that HCE, cyclo(Val-Pro), cyclo(Phe-Phe), cyclo(His-Pro), cyclo(Leu-Lys), and arginine exerted stronger anti-inflammatory and antioxidant effects among the 12 bioactives in amyloid β (Aβ)-treated HT-22 cells. Further transcriptome sequencing and polymerase chain reaction (PCR) array analysis showed that these bioactives participated in different signaling pathways, and cyclo(Val-Pro) was identified as the most potent cyclic dipeptide. In addition, the antiapoptotic and neuroprotective effect of cyclo(Val-Pro) was partly regulated by the activation of PI3K/AKT and AMPK pathways, and the inhibition of these pathways abolished the effect of cyclo(Val-Pro). Moreover, cyclo(Val-Pro) enhanced cognitive function and neurogenesis and alleviated neuroinflammation and oxidative stress in middle-aged mice, with an effect similar to HCE. Hippocampal transcriptome analysis further revealed that HCE and cyclo(Val-Pro) significantly enriched the neuroactive ligand-receptor interaction pathway, verified by enhanced neurotransmitter levels and upregulated neurotransmitter receptor-related gene expression. Therefore, the mechanism of cyclo(Val-Pro) on neural function might be associated with PI3K/AKT and AMPK pathway-mediated antiapoptotic effect and neurogenesis and the activation of the neurotransmitter-receptor pathway.
Collapse
Affiliation(s)
- Yinhua Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Zhaorong Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Fen Zhuge
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
| | - Kexin Zhou
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Liujie Zheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Xinyang Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Sisi Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Ou Fu
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| |
Collapse
|
17
|
Bahorik AL, Hoang TD, Jacobs DR, Levine DA, Yaffe K. Association of Changes in C-Reactive Protein Level Trajectories Through Early Adulthood With Cognitive Function at Midlife: The CARDIA Study. Neurology 2024; 103:e209526. [PMID: 38959452 PMCID: PMC11226328 DOI: 10.1212/wnl.0000000000209526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/02/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Late-life inflammation has been linked to dementia risk and preclinical cognitive decline, but less is known about early adult inflammation and whether this could influence cognition in midlife. We aimed to identify inflammation levels through early adulthood and determine association of these trajectories with midlife cognition. METHODS We used data from the Coronary Artery Risk Development in Young Adults study to identify inflammation trajectories (C-reactive protein [CRP] level <10 mg/L) over 18 years through early adulthood (age range 24-58) in latent class analysis and to assess associations with cognition 5 years after the last CRP measurement (age range 47-63). Six cognitive domains were evaluated from tests of verbal memory, processing speed, executive function, verbal and category fluency, and global cognition; poor cognitive performance was defined as a decline of ≥1 SD less than the mean on each domain. The primary outcome was poor cognitive performance. Logistic regression was used to adjust for demographics, smoking, alcohol use, physical activity, and APOE 4 status. RESULTS Among 2,364 participants, the mean (SD) age was 50.2 (3.5) years; 55% were female, and 57% were White. Three CRP trajectories emerged over 18 years: lower stable (45%), moderate/increasing (16%), and consistently higher (39%). Compared with lower stable CRP, both consistently higher (adjusted odds ratio [aOR] 1.67, 95% CI 1.23-2.26) and moderately/increasing (aOR 2.04, 95% CI 1.40-2.96) CRP had higher odds of poor processing speed; consistently higher CRP additionally had higher odds of poor executive function (aOR 1.36, 95% CI 1.00-1.88). For memory (moderately/increasing aOR 1.36, 95% CI 1.00-1.88; consistently higher aOR 1.18, 95% CI 0.90-1.54), letter fluency (moderately/increasing aOR 1.00, 95% CI 0.69-1.43; consistently higher aOR 1.05, 95% CI 0.80-1.39), category fluency (moderately/increasing aOR 1.16, 95% CI 0.82-1.63; consistently higher aOR 1.11, 95% CI 0.85-1.45), or global cognition (moderately/increasing aOR 1.16, 95% CI 0.82-1.63; consistently higher aOR 1.11, 95% CI 0.85-1.45), no association was observed. DISCUSSION Consistently higher or moderate/increasing inflammation starting in early adulthood may lead to worse midlife executive function and processing speed. Study limitations include selection bias due to loss to follow-up and reliance on CRP as the only inflammatory marker. Inflammation is important for cognitive aging and may begin much earlier than previously known.
Collapse
Affiliation(s)
- Amber L Bahorik
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| | - Tina D Hoang
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| | - David R Jacobs
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| | - Deborah A Levine
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| | - Kristine Yaffe
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| |
Collapse
|
18
|
Zhang S, Xiao Y, Cheng Y, Ma Y, Liu J, Li C, Shang H. Associations of sugar intake, high-sugar dietary pattern, and the risk of dementia: a prospective cohort study of 210,832 participants. BMC Med 2024; 22:298. [PMID: 39020335 PMCID: PMC11256505 DOI: 10.1186/s12916-024-03525-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Limited evidence demonstrated the potential relationship between dietary sugar intake and dementia. This association demands further clarification in a large-scale population. METHODS A total of 210,832 participants from the UK Biobank cohort were included in this prospective cohort study. Absolute and relative sugar intake and high-sugar dietary scores were utilized to reflect dietary sugar intake. Absolute sugar intake was identified by the Oxford WebQ in the UK Biobank. Relative sugar intake was calculated by dividing the absolute sugar intake by total diet energy. High-sugar dietary pattern was identified using the method of reduced rank regression. Cox proportional hazards regression analyses and restricted cubic splines were performed to examine the longitudinal associations between dietary sugar intake and all-cause dementia and its main subtype, Alzheimer's disease. Explorative mediation analyses were conducted to explore underlying mechanisms. RESULTS Increased absolute sugar intake (g/day) was significantly associated with a higher risk of all-cause dementia (HR = 1.003, [95%CI: 1.002-1.004], p < 0.001) and Alzheimer's disease (1.002, [1.001-1.004], 0.005). Relative sugar intake (%g/kJ/day) also demonstrated significant associations with all-cause dementia (1.317, [1.173-1.480], p < 0.001) and Alzheimer's disease (1.249, [1.041-1.500], 0.017), while the high-sugar dietary score was only significantly associated with a higher risk of all-cause dementia (1.090, [1.045-1.136], p < 0.001). In addition, both sugar intake and high-sugar dietary score demonstrated significant non-linear relationships with all-cause dementia and Alzheimer's disease (all p values for non-linearity < 0.05). CONCLUSIONS Our study provided evidence that excessive sugar intake was associated with dementia. Controlling the excess consumption of dietary sugar may be of great public health implications for preventing dementia.
Collapse
Affiliation(s)
- Sirui Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Xiao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yangfan Cheng
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuanzheng Ma
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiyong Liu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, 610041, China
- School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Neurodegenerative Disorders, National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
Soria-Contreras DC, Liu J, Lawn RB, Wang S, Purdue-Smithe A, Grodstein F, Oken E, Chavarro JE. Lifetime History of Low Birth Weight Delivery and Cognitive Function in Middle-Aged Parous Women. Neurology 2024; 103:e209504. [PMID: 38865681 DOI: 10.1212/wnl.0000000000209504] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Pregnancy outcomes such as low birth weight (LBW) delivery may reflect vascular or metabolic dysfunction in mothers and presage future cognitive impairment and dementia. However, the evidence is currently limited. Our objective was to examine the extent to which a lifetime history of LBW delivery was associated with cognitive function in parous middle-aged women. METHODS We studied participants from the Nurses' Health Study II, an ongoing longitudinal cohort of female nurses enrolled in 1989. In 2009, participants completed a reproductive history questionnaire. Participants who completed at least one of 2 post-traumatic stress disorder questionnaires were invited to participate in a cognition substudy with 2 waves of baseline data collection (2014 or 2018). We restricted the analysis to participants with one valid cognitive assessment who reported ≥1 birth at 18 years and older. We defined LBW delivery history as having delivered offspring with a birth weight <2,500 g (<5.5 lbs) in any pregnancy. The outcome was a single assessment of cognitive function evaluated with the self-administered Cogstate Brief Battery. The battery comprises 4 tasks, which we used to create 2 composite z-scores measuring psychomotor speed/attention and learning/working memory (higher z-scores = better cognitive function). We used multivariable linear regression models. RESULTS The analysis included 15,323 participants with a mean age of 62 (standard deviation: 4.9 years) at cognitive assessment. Among them, 1,224 (8%) had a history of LBW delivery. After adjusting for age at cognitive assessment, race, and ethnicity, participants' education, wave of baseline cognitive assessment, socioeconomic status, and prepregnancy characteristics, women with a history of LBW delivery had lower z-scores in the psychomotor speed/attention (β, -0.06; 95% CI -0.12 to -0.01) and learning/working memory (β, -0.05; 95% CI -0.09 to -0.01) composites than parous women without a history of LBW delivery. We observed a gradient of lower z-scores with an increasing number of LBW deliveries. DISCUSSION History of LBW delivery may be marker of future poorer cognition. If confirmed, our findings support future investigations into the value of early preventive efforts targeting women with a history of LBW delivery to reduce the burden of cognitive impairment in women.
Collapse
Affiliation(s)
- Diana C Soria-Contreras
- From the Departments of Nutrition (D.C.S.-C., S.W., E.O., J.E.C.) and Epidemiology (J.L., R.B.L., J.E.C.), Harvard T.H. Chan School of Public Health, Boston; Division of Women's Health (A.P.-S.) and Channing Division of Network Medicine, Department of Medicine (J.E.C.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine (E.O.), Harvard Medical School, Boston; and Harvard Pilgrim Health Care Institute (E.O.), Boston, MA
| | - Jiaxuan Liu
- From the Departments of Nutrition (D.C.S.-C., S.W., E.O., J.E.C.) and Epidemiology (J.L., R.B.L., J.E.C.), Harvard T.H. Chan School of Public Health, Boston; Division of Women's Health (A.P.-S.) and Channing Division of Network Medicine, Department of Medicine (J.E.C.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine (E.O.), Harvard Medical School, Boston; and Harvard Pilgrim Health Care Institute (E.O.), Boston, MA
| | - Rebecca B Lawn
- From the Departments of Nutrition (D.C.S.-C., S.W., E.O., J.E.C.) and Epidemiology (J.L., R.B.L., J.E.C.), Harvard T.H. Chan School of Public Health, Boston; Division of Women's Health (A.P.-S.) and Channing Division of Network Medicine, Department of Medicine (J.E.C.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine (E.O.), Harvard Medical School, Boston; and Harvard Pilgrim Health Care Institute (E.O.), Boston, MA
| | - Siwen Wang
- From the Departments of Nutrition (D.C.S.-C., S.W., E.O., J.E.C.) and Epidemiology (J.L., R.B.L., J.E.C.), Harvard T.H. Chan School of Public Health, Boston; Division of Women's Health (A.P.-S.) and Channing Division of Network Medicine, Department of Medicine (J.E.C.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine (E.O.), Harvard Medical School, Boston; and Harvard Pilgrim Health Care Institute (E.O.), Boston, MA
| | - Alexandra Purdue-Smithe
- From the Departments of Nutrition (D.C.S.-C., S.W., E.O., J.E.C.) and Epidemiology (J.L., R.B.L., J.E.C.), Harvard T.H. Chan School of Public Health, Boston; Division of Women's Health (A.P.-S.) and Channing Division of Network Medicine, Department of Medicine (J.E.C.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine (E.O.), Harvard Medical School, Boston; and Harvard Pilgrim Health Care Institute (E.O.), Boston, MA
| | - Francine Grodstein
- From the Departments of Nutrition (D.C.S.-C., S.W., E.O., J.E.C.) and Epidemiology (J.L., R.B.L., J.E.C.), Harvard T.H. Chan School of Public Health, Boston; Division of Women's Health (A.P.-S.) and Channing Division of Network Medicine, Department of Medicine (J.E.C.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine (E.O.), Harvard Medical School, Boston; and Harvard Pilgrim Health Care Institute (E.O.), Boston, MA
| | - Emily Oken
- From the Departments of Nutrition (D.C.S.-C., S.W., E.O., J.E.C.) and Epidemiology (J.L., R.B.L., J.E.C.), Harvard T.H. Chan School of Public Health, Boston; Division of Women's Health (A.P.-S.) and Channing Division of Network Medicine, Department of Medicine (J.E.C.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine (E.O.), Harvard Medical School, Boston; and Harvard Pilgrim Health Care Institute (E.O.), Boston, MA
| | - Jorge E Chavarro
- From the Departments of Nutrition (D.C.S.-C., S.W., E.O., J.E.C.) and Epidemiology (J.L., R.B.L., J.E.C.), Harvard T.H. Chan School of Public Health, Boston; Division of Women's Health (A.P.-S.) and Channing Division of Network Medicine, Department of Medicine (J.E.C.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Rush Alzheimer's Disease Center (F.G.), Rush University Medical Center, Chicago, IL; Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine (E.O.), Harvard Medical School, Boston; and Harvard Pilgrim Health Care Institute (E.O.), Boston, MA
| |
Collapse
|
20
|
Oken BS, Kaplan J, Klee D, Gallegos AM. Contributions of loneliness to cognitive impairment and dementia in older adults are independent of other risk factors and Alzheimer's pathology: a narrative review. Front Hum Neurosci 2024; 18:1380002. [PMID: 38873650 PMCID: PMC11169707 DOI: 10.3389/fnhum.2024.1380002] [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: 01/31/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Loneliness significantly contributes to cognitive impairment and dementia in older adults. Loneliness is a distressing feeling resulting from a perceived lack of social connection (i.e., a discrepancy between desired and actual social relationships), while social isolation is a related term that can be defined by number and type of social relationships. Importantly, loneliness is distinct from social isolation in that it is associated with a distressing self-perception. The primary focus of this narrative review is the impact of chronic loneliness on cognitive impairment and dementia among older adults. Loneliness has a significant association with many factors that are related to worse cognition, and therefore we include discussion on health, mental health, as well as the physiological effects of loneliness, neuropathology, and potential treatments. Loneliness has been shown to be related to development of dementia with a hazard ratio (HR) risk comparable to having a single APOE4 gene. The relationship of dementia to loneliness appears to be at least partially independent of other known dementia risk factors that are possibly associated with loneliness, such as depression, educational status, social isolation, and physical activity. Episodic memory is not consistently impacted by loneliness, which would be more typically impaired if the mild cognitive impairment (MCI) or dementia was due to Alzheimer's disease (AD) pathology. In addition, the several longitudinal studies that included neuropathology showed no evidence for a relationship between loneliness and AD neuropathology. Loneliness may decrease resilience, or produce greater cognitive change associated with the same level of AD neuropathology. Intervention strategies to decrease loneliness in older adults have been developed but need to consider key treatment targets beyond social isolation. Loneliness needs to be assessed in all studies of cognitive decline in elders, since it significantly contributes to the variance of cognitive function. It will be useful to better define the underlying mechanism of loneliness effects on cognition to determine if it is similar to other psychological factors related to excessive stress reactivity, such as neuroticism or even depression, which are also associated with cognitive decline. It is important from a health perspective to develop better strategies to decrease loneliness in older adults.
Collapse
Affiliation(s)
- Barry S. Oken
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Josh Kaplan
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
| | - Daniel Klee
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
| | - Autumn M. Gallegos
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
21
|
Sehgal R, Markov Y, Qin C, Meer M, Hadley C, Shadyab AH, Casanova R, Manson JE, Bhatti P, Crimmins EM, Hägg S, Assimes TL, Whitsel EA, Higgins-Chen AT, Levine M. Systems Age: A single blood methylation test to quantify aging heterogeneity across 11 physiological systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.13.548904. [PMID: 37503069 PMCID: PMC10370047 DOI: 10.1101/2023.07.13.548904] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Individuals, organs, tissues, and cells age in diverse ways throughout the lifespan. Epigenetic clocks attempt to quantify differential aging between individuals, but they typically summarize aging as a single measure, ignoring within-person heterogeneity. Our aim was to develop novel systems-based methylation clocks that, when assessed in blood, capture aging in distinct physiological systems. We combined supervised and unsupervised machine learning methods to link DNA methylation, system-specific clinical chemistry and functional measures, and mortality risk. This yielded a panel of 11 system-specific scores- Heart, Lung, Kidney, Liver, Brain, Immune, Inflammatory, Blood, Musculoskeletal, Hormone, and Metabolic. Each system score predicted a wide variety of outcomes, aging phenotypes, and conditions specific to the respective system. We also combined the system scores into a composite Systems Age clock that is predictive of aging across physiological systems in an unbiased manner. Finally, we showed that the system scores clustered individuals into unique aging subtypes that had different patterns of age-related disease and decline. Overall, our biological systems based epigenetic framework captures aging in multiple physiological systems using a single blood draw and assay and may inform the development of more personalized clinical approaches for improving age-related quality of life.
Collapse
|
22
|
Rani A, Bean L, Budamagunta V, Kumar A, Foster TC. Failure of senolytic treatment to prevent cognitive decline in a female rodent model of aging. Front Aging Neurosci 2024; 16:1384554. [PMID: 38813533 PMCID: PMC11133672 DOI: 10.3389/fnagi.2024.1384554] [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: 02/09/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
There are sex differences in vulnerability and resilience to the stressors of aging and subsequent age-related cognitive decline. Cellular senescence occurs as a response to damaging or stress-inducing stimuli. The response includes a state of irreversible growth arrest, the development of a senescence-associated secretory phenotype, and the release of pro-inflammatory cytokines associated with aging and age-related diseases. Senolytics are compounds designed to eliminate senescent cells. Our recent work indicates that senolytic treatment preserves cognitive function in aging male F344 rats. The current study examined the effect of senolytic treatment on cognitive function in aging female rats. Female F344 rats (12 months) were treated with dasatinib (1.2 mg/kg) + quercetin (12 mg/kg) or ABT-263 (12 mg/kg) or vehicle for 7 months. Examination of the estrus cycle indicated that females had undergone estropause during treatment. Senolytic treatment may have increased sex differences in behavioral stress responsivity, particularly for the initial training on the cued version of the watermaze. However, pre-training on the cue task reduced stress responsivity for subsequent spatial training and all groups learned the spatial discrimination. In contrast to preserved memory observed in senolytic-treated males, all older females exhibited impaired episodic memory relative to young (6-month) females. We suggest that the senolytic treatment may not have been able to compensate for the loss of estradiol, which can act on aging mechanisms for anxiety and memory independent of cellular senescence.
Collapse
Affiliation(s)
- Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Linda Bean
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Vivekananda Budamagunta
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
- Genetics and Genomics Graduate Program, Genetics Institute, University of Florida, Gainesville, FL, United States
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Thomas C. Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
- Genetics and Genomics Graduate Program, Genetics Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
23
|
Sánchez-Ortí JV, Balanzá-Martínez V, Correa-Ghisays P, Selva-Vera G, Vila-Francés J, Magdalena-Benedito R, San-Martin C, Victor VM, Escribano-Lopez I, Hernandez-Mijares A, Vivas-Lalinde J, Crespo-Facorro B, Tabarés-Seisdedos R. Inflammation and weight change related to neurocognitive and functional impairment in diabetes and psychiatric disorders. SPANISH JOURNAL OF PSYCHIATRY AND MENTAL HEALTH 2024:S2950-2853(24)00030-9. [PMID: 38740330 DOI: 10.1016/j.sjpmh.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 04/24/2024] [Accepted: 05/05/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION Obesity is a global pandemic associated with various cardio-metabolic and psychiatric disorders. Neurocognitive and functional deficits have been associated with several somatic and psychiatric disorders. Adiposity-related inflammation has recently emerged as a key risk factor for neurocognitive and functional impairments. This prospective transdiagnostic study aimed to investigate the role of adiposity-related inflammatory markers in neurocognitive and functional outcomes associated with weight changes. METHODS Peripheral blood inflammatory and oxidative stress biomarkers and neurocognitive and functional performance were assessed twice over 1 year in 165 individuals, including 30 with schizophrenia, 42 with bipolar disorder, 35 with major depressive disorder, 30 with type 2 diabetes mellitus (T2DM), and 28 healthy controls. Participants were stratified by body mass index into categories of type 2 obesity (T2OB; n=30), type 1 obesity (T1OB; n=42), overweight (OW; n=53), and average weight (NW; n=40). Mixed one-way analysis of covariance and linear and binary logistic regression analyses were performed. RESULTS Compared with NW, T2OB and T1OB were significantly associated with impaired neurocognitive and functional performance (p<0.01; η2p=0.06-0.12) and higher levels of C-reactive protein and platelets (PLT) (p<0.01; η2p=0.08-0.16), with small-to-moderate effect sizes. IL-6, IL-10, and PLT were key factors for detecting significant weight changes in T1OB and T2OB over time. Regression models revealed that inflammatory and oxidative stress biomarkers and cellular adhesion molecules were significantly associated with neurocognitive and functional performance (p<0.05). DISCUSSION Obesity is characterized by neurocognitive and functional impairments alongside low-grade systemic inflammation. Adiposity-related inflammatory biomarkers may contribute to neurocognitive and functional decline in individuals with T2DM and psychiatric disorders. Our data suggest that these biomarkers facilitate the identification of specific subgroups of individuals at higher risk of developing obesity.
Collapse
Affiliation(s)
- Joan Vicent Sánchez-Ortí
- INCLIVA - Health Research Institute, Valencia, Spain; Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Faculty of Psychology, University of Valencia, Valencia, Spain
| | - Vicent Balanzá-Martínez
- INCLIVA - Health Research Institute, Valencia, Spain; Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain; Mental Health Unit of Catarroja, Valencia, Spain
| | - Patricia Correa-Ghisays
- INCLIVA - Health Research Institute, Valencia, Spain; Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Faculty of Psychology, University of Valencia, Valencia, Spain; Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain
| | - Gabriel Selva-Vera
- INCLIVA - Health Research Institute, Valencia, Spain; Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain
| | - Joan Vila-Francés
- IDAL - Intelligent Data Analysis Laboratory, University of Valencia, Valencia, Spain
| | | | - Constanza San-Martin
- TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Department of Physiotherapy, University of Valencia, Valencia, Spain
| | - Víctor M Victor
- INCLIVA - Health Research Institute, Valencia, Spain; Service of Endocrinology and Nutrition, University Hospital Dr. Peset, Spain; Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Department of Physiology, University of Valencia, Valencia, Spain
| | | | | | | | - Benedicto Crespo-Facorro
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; Department of Psychiatry, Faculty of Medicine, University of Sevilla, HU Virgen del Rocío IBIS, Spain
| | - Rafael Tabarés-Seisdedos
- INCLIVA - Health Research Institute, Valencia, Spain; Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain.
| |
Collapse
|
24
|
Patel R, Cosentino S, Zheng EZ, Schupf N, Barral S, Feitosa M, Andersen SL, Sebastiani P, Ukraintseva S, Christensen K, Zmuda J, Thyagarajan B, Gu Y. Systemic inflammation in relation to exceptional memory in the Long Life Family Study (LLFS). Brain Behav Immun Health 2024; 37:100746. [PMID: 38476338 PMCID: PMC10925922 DOI: 10.1016/j.bbih.2024.100746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Background and objectives We previously found a substantial familial aggregation of healthy aging phenotypes, including exceptional memory (EM) in long-lived persons. In the current study, we aim to assess whether long-lived families with EM and without EM (non-EM) differ in systemic inflammation status and trajectory. Methods The current study included 4333 participants of the multi-center Long Life Family Study (LLFS). LLFS families were classified as EM (556 individuals from 28 families) or non-EM (3777 individuals from 416 families), with 2 or more offspring exhibiting exceptional memory performance (i.e. having baseline composite z-score representing immediate and delayed story memory being 1.5 SD above the mean in the nondemented offspring sample) considered as EM. Blood samples from baseline were used to measure inflammatory biomarkers including total white blood cell (WBC) and its subtypes (neutrophils, lymphocytes, monocytes) count, platelet count, high sensitivity C-reactive protein, and interleukin-6. Generalized linear models were used to examine cross-sectional differences in inflammatory biomarkers at baseline. In a sub-sample of 2227 participants (338 subjects from 24 EM families and 1889 from 328 non-EM families) with repeated measures of immune cell counts, we examined whether the rate of biomarker change differed between EM and non-EM families. All models were adjusted for family size, relatedness, age, sex, education, field center, APOE genotype, and body mass index. Results LLFS participants from EM families had a marginally higher monocyte count at baseline (b = 0.028, SE = 0.0110, p = 0.010) after adjusting for age, sex, education, and field site, particularly in men (p < 0.0001) but not in women (p = 0.493) (p-interaction = 0.003). Over time, monocyte counts increased (p < 0.0001) in both EM and non-EM families, while lymphocytes and platelet counts decreased over time in the non-EM families (p < 0.0001) but not in the EM families. After adjusting for multiple variables, there was no significant difference in biomarker change over time between the EM and non-EM families. Discussion Compared with non-EM families, EM families had significantly higher monocyte count at baseline but had similar change over time. Our study suggests that differences in monocyte counts may be a pathway through which EM emerges in some long-lived families, especially among men.
Collapse
Affiliation(s)
- Ruhee Patel
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Stephanie Cosentino
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Esther Zhiwei Zheng
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Nicole Schupf
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Sandra Barral
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Mary Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Stacy L. Andersen
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, 02111, USA
| | | | - Kaare Christensen
- Epidemiology, Biostatistics and Biodemography, University of Southern Denmark, 5230, Odense, Denmark
| | - Joseph Zmuda
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Yian Gu
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Long Life Family Study (LLFS)
- Cognitive Neuroscience Division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, 02111, USA
- Social Sciences Research Institute, Duke University, Durham, NC, 27705, USA
- Epidemiology, Biostatistics and Biodemography, University of Southern Denmark, 5230, Odense, Denmark
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
25
|
Muresan S, Slevin M. C-reactive Protein: An Inflammatory Biomarker and a Predictor of Neurodegenerative Disease in Patients With Inflammatory Bowel Disease? Cureus 2024; 16:e59009. [PMID: 38665135 PMCID: PMC11045161 DOI: 10.7759/cureus.59009] [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] [Accepted: 04/25/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammatory bowel disease (IBD) refers to two chronic conditions of the digestive tract: ulcerative colitis (UC) and Crohn's disease (CD), representing a progressive inflammatory process that mainly occurs in the gut, with frequent extra-intestinal manifestations. Even if remission is periodically obtained for some patients, the histological activity and digestive symptoms may continue, maintaining a persistent systemic inflammation that could induce further extra-intestinal complications and contribute to the development of neurodegenerative disease. C-reactive protein (CRP) is an acute-phase reactant that is widely accepted as a dominant serum biomarker in IBD. CRP consequently activates the complement cascade, supports the release of pro-inflammatory cytokines, and the clearance of microbial pathogens. All these processes facilitate further processes, including atherosclerosis and hypercoagulability, alteration of the intestinal microbiota, and the increased permeability of the intestinal barrier for neurotoxic substances produced by gut microorganisms, due to the presence of a high level of lipopolysaccharides. For IBD, the connection between intestinal inflammation and central nervous system inflammation could be explained through the activity of the vagus nerve, a carrier of cytokines, CRP, and toxic materials to the brain, potentially inducing vascular lesions and damage of the glial vascular unit, with further risk for degeneration within the central nervous system. CRP is a key marker for IBD pathogenesis and is able to dissociate into its monomeric form, mCRP, on contact with activated cell and tissue components via the systemic circulation. We hypothesize that the chronic inflammatory process within IBD could initiate neuroinflammation and neurodegeneration, and therefore, further investigation of the significance of chronically raised plasma of CRP and mCRP in patients with IBD is warranted, as it may represent a critical predictive factor associated with a later neurodegenerative risk. Any future initiative aimed at pharmacologic modulation of CRP (e.g., blocking CRP-mCRP dissociation), could represent a new therapeutic approach protecting against intestinal inflammation and concomitantly reducing the risk of neuroinflammation, neurodegeneration, and cognitive decline.
Collapse
Affiliation(s)
- Simona Muresan
- Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, Targu Mures, ROU
| | - Mark Slevin
- Center for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Sciences and Technology, Targu Mures, ROU
| |
Collapse
|
26
|
Brown GC, Heneka MT. The endotoxin hypothesis of Alzheimer's disease. Mol Neurodegener 2024; 19:30. [PMID: 38561809 PMCID: PMC10983749 DOI: 10.1186/s13024-024-00722-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: 11/17/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Lipopolysaccharide (LPS) constitutes much of the surface of Gram-negative bacteria, and if LPS enters the human body or brain can induce inflammation and act as an endotoxin. We outline the hypothesis here that LPS may contribute to the pathophysiology of Alzheimer's disease (AD) via peripheral infections or gut dysfunction elevating LPS levels in blood and brain, which promotes: amyloid pathology, tau pathology and microglial activation, contributing to the neurodegeneration of AD. The evidence supporting this hypothesis includes: i) blood and brain levels of LPS are elevated in AD patients, ii) AD risk factors increase LPS levels or response, iii) LPS induces Aβ expression, aggregation, inflammation and neurotoxicity, iv) LPS induces TAU phosphorylation, aggregation and spreading, v) LPS induces microglial priming, activation and neurotoxicity, and vi) blood LPS induces loss of synapses, neurons and memory in AD mouse models, and cognitive dysfunction in humans. However, to test the hypothesis, it is necessary to test whether reducing blood LPS reduces AD risk or progression. If the LPS endotoxin hypothesis is correct, then treatments might include: reducing infections, changing gut microbiome, reducing leaky gut, decreasing blood LPS, or blocking LPS response.
Collapse
Affiliation(s)
- Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
27
|
Gunawan PY, Gunawan PA, Hariyanto TI. Risk of Dementia in Patients with Diabetes Using Sodium-Glucose Transporter 2 Inhibitors (SGLT2i): A Systematic Review, Meta-Analysis, and Meta-Regression. Diabetes Ther 2024; 15:663-675. [PMID: 38340279 PMCID: PMC10942948 DOI: 10.1007/s13300-024-01538-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
INTRODUCTION Dementia is quite prevalent and among the leading causes of death worldwide. According to earlier research, diabetes may increase the possibility of developing dementia. However, the association between antidiabetic agents and dementia is not yet clear. This investigation examines the association between the use of sodium-glucose transporter 2 inhibitors (SGLT2i) and the risk of dementia in patients with diabetes. METHODS Up to April 18, 2023, four databases-Europe PMC, Medline, Scopus, and Cochrane Library-were searched for relevant literature. We included all studies that examine dementia risk in adults with diabetes who use SGLT2i. Random-effect models were used to compute the outcomes in this investigation, producing pooled odds ratios (OR) with 95% confidence intervals (CI). RESULTS Pooled data from seven observational studies revealed that SGLT2i use was linked to a lower risk of dementia in people with diabetes (OR 0.45, 95% CI 0.34-0.61; p < 0.00001, I2 = 97%). The reduction in the risk of dementia due to SGLT2i's neuroprotective effect was only significantly affected by dyslipidemia (p = 0.0004), but not by sample size (p = 0.2954), study duration (p = 0.0908), age (p = 0.0805), sex (p = 0.5058), hypertension (p = 0.0609), cardiovascular disease (p = 0.1619), or stroke (p = 0.2734). CONCLUSIONS According to this research, taking SGLT2i reduces the incidence of dementia in people with diabetes by having a beneficial neuroprotective impact. Randomized controlled trials (RCTs) are still required in order to verify the findings of our research.
Collapse
Affiliation(s)
- Pricilla Yani Gunawan
- Department of Neurology, Faculty of Medicine, Pelita Harapan University, Boulevard Jendral Sudirman Street, Karawaci, Tangerang, 15811, Indonesia.
| | - Paskalis Andrew Gunawan
- Division of Geriatric Medicine, Department of Internal Medicine, Faculty of Medicine, Tarumanegara University, Jakarta, 11440, Indonesia
| | | |
Collapse
|
28
|
Mishra SP, Jain S, Wang B, Wang S, Miller BC, Lee JY, Borlongan CV, Jiang L, Pollak J, Taraphder S, Layden BT, Rane SG, Yadav H. Abnormalities in microbiota/butyrate/FFAR3 signaling in aging gut impair brain function. JCI Insight 2024; 9:e168443. [PMID: 38329121 PMCID: PMC10967378 DOI: 10.1172/jci.insight.168443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 12/08/2023] [Indexed: 02/09/2024] Open
Abstract
Aging-related abnormalities in gut microbiota are associated with cognitive decline, depression, and anxiety, but underlying mechanisms remain unstudied. Here, our study demonstrated that transplanting old gut microbiota to young mice induced inflammation in the gut and brain coupled with cognitive decline, depression, and anxiety. We observed diminished mucin formation and increased gut permeability ("leaky gut") with a reduction in beneficial metabolites like butyrate because of decline in butyrate-producing bacteria in the aged gut microbiota. This led to suppressed expression of butyrate receptors, free fatty acid receptors 2 and 3 (FFAR2/3). Administering butyrate alleviated inflammation, restored mucin expression and gut barriers, and corrected brain dysfunction. Furthermore, young mice with intestine-specific loss of FFAR2/3 exhibited gut and brain abnormalities akin to those in older mice. Our results demonstrate that reduced butyrate-producing bacteria in aged gut microbiota result in low butyrate levels and reduced FFAR2/3 signaling, leading to suppressed mucin formation that increases gut permeability, inflammation, and brain abnormalities. These findings underscore the significance of butyrate-FFAR2/3 agonism as a potential strategy to mitigate aged gut microbiota-induced detrimental effects on gut and brain health in older adults.
Collapse
Affiliation(s)
- Sidharth P. Mishra
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Shalini Jain
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Bo Wang
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, Florida, USA
| | - Shaohua Wang
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Brandi C. Miller
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Jea Y. Lee
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
| | - Lin Jiang
- Natural Sciences Division, New College of Florida, Sarasota, Florida, USA
| | - Julie Pollak
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, Florida, USA
| | - Subhash Taraphder
- Department of Animal Genetics and Breeding, West Bengal University of Animal & Fishery Sciences, Kolkata, India
| | - Brian T. Layden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Sushil G. Rane
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Hariom Yadav
- USF Center for Microbiome Research
- Department of Neurosurgery and Brain Repair, and
- Center for Excellence of Aging and Brain Repair, University of South Florida (USF) Morsani College of Medicine, Tampa, Florida, USA
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, USF Morsani College of Medicine, Tampa, Florida, USA
| |
Collapse
|
29
|
Merten N, Fischer ME, Pinto AA, Chappell RJ, Schubert CR. Lifestyle and factors of vascular and metabolic health and inflammation are associated with sensorineural-neurocognitive aging in older adults. FRONTIERS IN EPIDEMIOLOGY 2024; 3:1299587. [PMID: 38455939 PMCID: PMC10910988 DOI: 10.3389/fepid.2023.1299587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/14/2023] [Indexed: 03/09/2024]
Abstract
This study's aim was to identify risk factors associated with sensorineural and neurocognitive function (brain aging) in older adults. In N = 1,478 Epidemiology of Hearing Loss Study participants (aged 64-100 years, 59% women), we conducted sensorineural and cognitive tests, which were combined into a summary measure using Principal Component Analysis (PCA). Participants with a PCA score <-1 standard deviation (SD) were considered to have brain aging. Incident brain aging was defined as PCA score <-1 SD at 5-year follow-up among participants who had a PCA score ≥-1 SD at baseline. Logistic regression and Poisson models were used to estimate associations between baseline risk factors of lifestyle, vascular and metabolic health, and inflammation and prevalent or incident brain aging, respectively. In an age-sex adjusted multivariable model, not consuming alcohol (odds ratio(OR) = 1.77, 95% confidence Interval (CI) = 1.18,2.66), higher interleukin-6 levels (OR = 1.30, 95% CI = 1.03,1.64), and depressive symptoms (OR = 2.44, 95% CI = 1.63,3.67) were associated with a higher odds of having brain aging, while higher education had protective effects (OR = 0.55, 95% CI = 0.33,0.94). A history of stroke, arterial stiffness, and obesity were associated with an increased risk of developing brain aging during the five years of follow-up. Lifestyle, vascular, metabolic and inflammatory factors were associated with brain aging in older adults, which adds to the evidence of shared pathways for sensorineural and neurocognitive declines in aging. Targeting these shared central processing etiological factors with interventions may lead to retention of better neurological function, benefiting multiple systems, i.e., hearing, smell, and cognition, ultimately helping older adults retain independence and higher quality of life longer.
Collapse
Affiliation(s)
- Natascha Merten
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Division of Geriatrics and Gerontology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Mary E. Fischer
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Aaron Alex Pinto
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Richard J. Chappell
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Carla R. Schubert
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
30
|
Ji D, Chen WZ, Zhang L, Zhang ZH, Chen LJ. Gut microbiota, circulating cytokines and dementia: a Mendelian randomization study. J Neuroinflammation 2024; 21:2. [PMID: 38178103 PMCID: PMC10765696 DOI: 10.1186/s12974-023-02999-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 12/16/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Some studies have shown that gut microbiota may be associated with dementia. However, the causal effects between gut microbiota and different types of dementia and whether cytokines act as a mediator remain unclear. METHODS Gut microbiota, cytokines, and five dementia types, including Alzheimer's disease (AD), frontotemporal dementia (FTD), dementia with Lewy body (DLB), vascular dementia (VD), and Parkinson's disease dementia (PDD) were identified from large-scale genome-wide association studies (GWAS) summary data. We used Mendelian randomization (MR) to investigate the causal relationships between gut microbiota, cytokines, and five types of dementia. Inverse variance weighting (IVW) was used as the main statistical method. In addition, we explored whether cytokines act as a mediating factor in the pathway from gut microbiota to dementia. RESULTS There were 20 positive and 16 negative causal effects between genetic liability in the gut microbiota and dementia. Also, there were five positive and four negative causal effects between cytokines and dementias. Cytokines did not act as mediating factors. CONCLUSIONS Gut microbiota and cytokines were causally associated with five types of dementia, and cytokines seemed not to be the mediating factors in the pathway from gut microbiota to dementia.
Collapse
Affiliation(s)
- Dong Ji
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, Anhui, China
| | - Wen-Zhu Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, Anhui, China
| | - Lei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, Anhui, China
| | - Zhi-Hua Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Li-Jian Chen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei, Anhui, China.
| |
Collapse
|
31
|
Garcia-Argibay M, Hiyoshi A, Montgomery S. Association between dementia risk and ulcerative colitis, with and without colectomy: a Swedish population-based register study. BMJ Open 2023; 13:e074110. [PMID: 38135306 DOI: 10.1136/bmjopen-2023-074110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2023] Open
Abstract
OBJECTIVE This study aims to investigate the association of ulcerative colitis (UC) with all-cause dementia and assess differences in those with and without a total colectomy. DESIGN, SETTING AND PARTICIPANTS This Swedish prospective register-based study comprised 4.8 million individuals aged at least 59 years between 1964 and 2018 with the linkage of several Swedish national registers. PRIMARY AND SECONDARY OUTCOME MEASURES Individuals with dementia were defined according to International Classification of Diseases diagnostic codes and Anatomical Therapeutic Classification codes for medication prescriptions. Fitting Cox hazards models, the risk of developing all-cause dementia in individuals with and without UC was estimated. Further, we compared the risk of all-cause dementia among those with and without a colectomy. RESULTS Among 4 821 488 individuals (52.6% females) followed for 84.1 million person-years between 1964 and 2018, the incidence rate of all-cause dementia was 63.90 (63.73-64.07) events per 10 000 person-years in individuals without UC, 94.80 (92.04-97.64) among those with UC, 95.01 (92.25-97.86) in those with UC but without colectomy and 63.42 (40.92-98.31) in those with UC and a colectomy. Adjusted Cox models showed an increased all-cause dementia risk in individuals with UC (HR 1.07, 95% CI 1.04 to 1.10). We found no differences between unexposed individuals and those with UC and a colectomy (HR 0.89, 95% CI 0.57 to 1.38). CONCLUSION The findings are consistent with previous evidence suggesting a slightly increased dementia risk among individuals with UC. This study provided no evidence of further risk increase of dementia among those who had a colectomy.
Collapse
Affiliation(s)
- Miguel Garcia-Argibay
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Orebro, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Ayako Hiyoshi
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Orebro, Sweden
- Department of Public Health Sciences, Stockholm University, Stockholm, Sweden
| | - Scott Montgomery
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Orebro, Sweden
- Department of Epidemiology and Public Health, University College London, London, UK
- Clinical Epidemiology Division, Department of Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
32
|
Zhu L, He A, Chen D, Dong X, Xiong X, Chen A. Cardiorespiratory fitness as a mediator between body fat rate and executive function in college students. Front Endocrinol (Lausanne) 2023; 14:1293388. [PMID: 38174333 PMCID: PMC10764023 DOI: 10.3389/fendo.2023.1293388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Purpose To examine whether body fat rate (BF%) is associated with cardiorespiratory fitness (CRF) and whether cardiorespiratory fitness (CRF) mediates the association between BF% and Executive function (EF) in young adults. Methods In this cross-sectional study, 226 college students were recruited from an university. Flanker, 2-back, and odder and shifting tasks were used to assess EF. The incremental cardiopulmonary exercise tests were performed, and maximal oxygen consumption was recorded during test. The body composition measuring instrument was used to evaluate the participants' BF%. Results The BF% of college students was negatively correlated with each EF, BF% was negatively correlated with CRF, and CRF was negatively correlated with EF (P< 0.001). Structural equation modeling (SEM) and simultaneous analysis of several groups were used to construct mediator model. The CRF of college students plays a partial mediating role between BF% and EF, and the mediating effect accounts for 48.8% of the total effect value. Sex has no moderate effect on the relationship between BF%, CRF, and EF. Conclusions College students with high BF% can improve their CRF by strengthening physical exercise, thereby indirectly improving their EF. Therefore, college students who have a higher body fat percentage should be compensated for engaging in physical exercise in order to enhance their CRF and mitigate the detrimental effects of obesity and overweight on EF.
Collapse
Affiliation(s)
- Lina Zhu
- College of Physical Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Aihong He
- College of Physical Education, Yangzhou Polytechnic College, Yangzhou, Jiangsu, China
| | - Dandan Chen
- College of Physical Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaoxiao Dong
- College of Physical Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xuan Xiong
- College of Physical Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Aiguo Chen
- College of Physical Education, Yangzhou University, Yangzhou, Jiangsu, China
- Nanjing Sport Institute, Nanjing, Jiangsu, China
| |
Collapse
|
33
|
Cacace R, Zhou L, Hendrickx Van de Craen E, Buist A, Hoogmartens J, Sieben A, Cras P, Vandenberghe R, De Deyn PP, Oehlrich D, De Bondt A, Engelborghs S, Moechars D, Van Broeckhoven C. Mutated Toll-like receptor 9 increases Alzheimer's disease risk by compromising innate immunity protection. Mol Psychiatry 2023; 28:5380-5389. [PMID: 37433968 PMCID: PMC11041692 DOI: 10.1038/s41380-023-02166-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 06/09/2023] [Accepted: 06/23/2023] [Indexed: 07/13/2023]
Abstract
The development of Alzheimer's disease (AD) involves central and peripheral immune deregulation. Gene identification and studies of AD genetic variants of peripheral immune components may aid understanding of peripheral-central immune crosstalk and facilitate new opportunities for therapeutic intervention. In this study, we have identified in a Flanders-Belgian family a novel variant p.E317D in the Toll-like receptor 9 gene (TLR9), co-segregating with EOAD in an autosomal dominant manner. In human, TLR9 is an essential innate and adaptive immune component predominantly expressed in peripheral immune cells. The p.E317D variant caused 50% reduction in TLR9 activation in the NF-κB luciferase assay suggesting that p.E317D is a loss-of-function mutation. Cytokine profiling of human PBMCs upon TLR9 activation revealed a predominantly anti-inflammatory response in contrast to the inflammatory responses from TLR7/8 activation. The cytokines released upon TLR9 activation suppressed inflammation and promoted phagocytosis of Aβ42 oligomers in human iPSC-derived microglia. Transcriptome analysis identified upregulation of AXL, RUBICON and associated signaling pathways, which may underline the effects of TLR9 signaling-induced cytokines in regulating the inflammatory status and phagocytic property of microglia. Our data suggest a protective role of TLR9 signaling in AD pathogenesis, and we propose that TLR9 loss-of-function may disrupt a peripheral-central immune crosstalk that promotes dampening of inflammation and clearance of toxic protein species, leading to the build-up of neuroinflammation and pathogenic protein aggregates in AD development.
Collapse
Affiliation(s)
- Rita Cacace
- Neurodegenerative Brain Diseases, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Lujia Zhou
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Elisabeth Hendrickx Van de Craen
- Neurodegenerative Brain Diseases, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology, University Hospital Antwerp, Edegem, Belgium
| | - Arjan Buist
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Julie Hoogmartens
- Neurodegenerative Brain Diseases, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anne Sieben
- Department of Pathology, University Hospital Antwerp, Edegem, Belgium
| | - Patrick Cras
- Department of Neurology, University Hospital Antwerp, Edegem, Belgium
- Institute Born-Bunge, Antwerp, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals Leuven, and Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Peter P De Deyn
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp, Antwerp, Belgium
| | - Daniel Oehlrich
- Discovery Sciences, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - An De Bondt
- Discovery Sciences, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Sebastiaan Engelborghs
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology, Universitair Ziekenhuis Brussel, and Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Diederik Moechars
- Department of Neuroscience, Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
34
|
Byrd DR, Martin DA, Joseph RP. Environmental, Sociocultural, Behavioral, and Biological Factors Associated with Cognitive Decline, Alzheimer's Disease, and Other Types of Dementia in Black Americans. CURR EPIDEMIOL REP 2023; 10:252-263. [PMID: 39206249 PMCID: PMC11349302 DOI: 10.1007/s40471-023-00337-9] [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] [Accepted: 09/18/2023] [Indexed: 09/04/2024]
Abstract
Purpose of Review This narrative review summarizes environmental, sociocultural, behavioral, and biological factors associated with cognitive decline and dementia in Black Americans. Recent Findings Variations in environmental factors (education, toxins) contribute to poor cognition in Blacks. Historical context, early-life educational experiences, and environmental exposures should be considered for addressing late-life cognitive disparities. Sociocultural (stress, discrimination, social networks, social activity, loneliness) and behavioral (depression, coping, health behaviors) factors can have positive and negative implications for cognitive aging. Given these factors are not consistently shown to play a role in maintaining cognition in Blacks, further examination is needed. Cardiometabolic conditions increase the risk of cognitive issues and are more common in Blacks; thus, examination of biological mechanisms (inflammation, vascular changes, etc.) warrants further study. Summary Future studies should explore the impact of education and segregation and identify mechanisms linking stress and discrimination to cognitive outcomes. Further, culturally tailored programs focused on preventative behaviors are needed to enhance health outcomes and reduce disparities.
Collapse
Affiliation(s)
- DeAnnah R. Byrd
- Edson College of Nursing and Health Innovation, Arizona State University, Health North, Suite 301, 550 N 3rd Street, Phoenix, AZ 85004, USA
- Center for Innovation in Healthy and Resilient Aging, Edson College of Nursing and Health Innovation, Arizona State University, 500 N 3rd St., Phoenix, AZ 85004, USA
| | - Danielle A. Martin
- Edson College of Nursing and Health Innovation, Arizona State University, Health North, Suite 301, 550 N 3rd Street, Phoenix, AZ 85004, USA
| | - Rodney P. Joseph
- Edson College of Nursing and Health Innovation, Arizona State University, Health North, Suite 301, 550 N 3rd Street, Phoenix, AZ 85004, USA
- Center for Health Promotion and Disease Prevention, Edson College of Nursing and Health Innovation, Arizona State University, 500 N 3rd St., Phoenix, AZ 85004, USA
| |
Collapse
|
35
|
Gentreau M, Rukh G, Miguet M, Clemensson LE, Alsehli AM, Titova OE, Schiöth HB. The Effects of Statins on Cognitive Performance Are Mediated by Low-Density Lipoprotein, C-Reactive Protein, and Blood Glucose Concentrations. J Gerontol A Biol Sci Med Sci 2023; 78:1964-1972. [PMID: 37431946 PMCID: PMC10613010 DOI: 10.1093/gerona/glad163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Indexed: 07/12/2023] Open
Abstract
Statins are widely used for cardiovascular disease prevention but their effects on cognition remain unclear. Statins reduce cholesterol concentration and have been suggested to provide both beneficial and detrimental effects. Our aim was to investigate the cross-sectional and longitudinal association between statin use and cognitive performance, and whether blood low-density lipoprotein, high-density lipoprotein, triglycerides, glucose, C-reactive protein, and vitamin D biomarkers mediated this association. We used participants from the UK biobank aged 40-69 without neurological and psychiatric disorders (n = 147 502 and n = 24 355, respectively). We performed linear regression to evaluate the association between statin use and cognitive performance and, mediation analysis to quantify the total, direct, indirect effects and the proportion meditated by blood biomarkers. Statin use was associated with lower cognitive performance at baseline (β = -0.40 [-0.53, -0.28], p = <.0001), and this association was mediated by low-density lipoprotein (proportion mediated = 51.4%, p = .002), C-reactive protein (proportion mediated = -11%, p = .006) and blood glucose (proportion mediated = 2.6%, p = .018) concentrations. However, statin use was not associated with cognitive performance, measured 8 years later (β = -0.003 [-0.11, 0.10], p = .96). Our findings suggest that statins are associated with lower short-term cognitive performance by lowering low-density lipoprotein and raising blood glucose concentrations, and better performance by lowering C-reactive protein concentrations. In contrast, statins have no effect on long-term cognition and remain beneficial in reducing cardiovascular risk factors.
Collapse
Affiliation(s)
- Mélissa Gentreau
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, University of Uppsala, Uppsala, Sweden
| | - Gull Rukh
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, University of Uppsala, Uppsala, Sweden
| | - Maud Miguet
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, University of Uppsala, Uppsala, Sweden
| | - Laura E Clemensson
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, University of Uppsala, Uppsala, Sweden
| | - Ahmed M Alsehli
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Olga E Titova
- Medical Epidemiology, Department of Surgical Sciences, University of Uppsala, Uppsala, Sweden
| | - Helgi B Schiöth
- Functional Pharmacology and Neuroscience, Department of Surgical Sciences, University of Uppsala, Uppsala, Sweden
| |
Collapse
|
36
|
Wu A, Sharrett AR, Folsom AR, Alonso A, Walker KA, Gottesman RF, Gross AL, Rawlings AM, Schneider ALC, Coresh J. Midlife Hemostasis Measures, 20-Year Cognitive Decline, and Incident Dementia. Neurology 2023; 101:e1697-e1707. [PMID: 37652701 PMCID: PMC10624500 DOI: 10.1212/wnl.0000000000207771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/27/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Blood concentrations of hemostatic factors affect thrombosis and bleeding diathesis and may contribute to cognitive impairment through modifiable vascular pathologies. Whether hemostasis, assessed in middle age, is associated with late-life cognitive impairment remains largely unknown in a community-dwelling population. METHODS Using data from 14,128 participants with cognitive function measurements in 1990-1992 from the Atherosclerosis Risk in Communities study, we assessed the associations of hemostasis measures with 20-year changes in cognitive performance and incident dementia. Activated partial thromboplastin time (aPTT) and level of fibrinogen, von Willebrand factor (VWF), factor VIII, factor VII, factor XI, d-dimer, and soluble thrombomodulin were measured in 1987-1989 or 1993-1995. Hemostasis measures were categorized into quintiles, with the lowest quintile indicating low coagulability. Cognitive performance was characterized using a combined z-score from 3 tests (that is, delayed word recall test [DWRT], digit symbol substitution [DSST], and word fluency test [WFT]), assessed in 1990-1992, 1996-1998, and 2011-2013. Dementia was determined either from in-person evaluations or using dementia surveillance through 2017. Mixed-effects models and Cox proportional hazards models were used to assess cognitive trajectories and risk of dementia, respectively. RESULTS Among 12,765 participants with hemostasis measures in 1987-1989, who were aged 47-70 years at the first cognitive assessment, we observed significant trends of shorter aPTT (p for trend <0.001; difference in 20-year cognitive decline for fifth vs first quintile [Q5 vs Q1]: -0.104 [95% CI -0.160 to -0.048]) and higher levels of factor VII (p < 0.002; Q5 vs Q1: -0.085 [-0.142, -0.028]) and factor VIII (p = 0.033; Q4 vs Q1: -0.055 [-0.111, -0.000]) with greater 20-year cognitive declines. The associations with the decline in DSST were stronger than those with the decline in WFT or DWRT. Consistently, shorter aPTT and higher factor VIII levels were associated with higher dementia risk with HRs for Q5 vs Q1 of 1.23 (95% CI 1.07 to 1.42) and 1.17 (1.01-1.36), respectively, and p for trend of 0.008 and 0.024, respectively. DISCUSSION Overall, our study found consistent trend associations of aPTT and factor VIII measured in midlife with cognitive decline and incident dementia over 20 years, likely driven by vascular pathologies.
Collapse
Affiliation(s)
- Aozhou Wu
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - A Richey Sharrett
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Aaron R Folsom
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Alvaro Alonso
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Keenan A Walker
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Rebecca F Gottesman
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Alden L Gross
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Andreea M Rawlings
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Andrea Lauren Christman Schneider
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Josef Coresh
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia.
| |
Collapse
|
37
|
Acevedo-Fontánez AI, Cvejkus RK, Zmuda JM, Kuipers AL, Barinas-Mitchell E, Sekikawa A, Wheeler V, Rosano C, Miljkovic I. Skeletal muscle adiposity is a novel risk factor for poor cognition in African Caribbean women. Obesity (Silver Spring) 2023; 31:2398-2406. [PMID: 37475604 PMCID: PMC10680092 DOI: 10.1002/oby.23816] [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: 12/05/2022] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 07/22/2023]
Abstract
OBJECTIVE Skeletal muscle adiposity (myosteatosis) is recognized as a major risk factor for cardiometabolic diseases, and it increases with aging. The relationship of myosteatosis with cognitive impairment is unknown. METHODS The association of calf myosteatosis (measured by computed tomography-derived skeletal muscle density; higher values indicate less myosteatosis) with cognitive function was examined among 626 African Caribbean women who were aged 40 to 84 years, a population highly vulnerable to increased myosteatosis. Cognition was assessed by the Digit Symbol Substitution Test (DSST), a test of information processing speed (higher scores indicate better performance). Linear regression was used to assess the association of muscle density with DSST. RESULTS Adjusting for age, education, muscle area, BMI, hypertension, diabetes, cardiovascular event history, lifestyle factors, lipid-lowering medication use, and menopausal status, a one-SD lower muscle density was associated with a 1.69-point lower DSST score (p = 0.002). BMI, diabetes, and hypertension interactions were not statistically significant, suggesting that the main association was not moderated by overall obesity or cardiometabolic diseases. CONCLUSIONS These findings suggest that greater myosteatosis is associated with slower information processing speed, an early indicator of cognitive impairment. Further studies are needed to establish this association in this and other populations using an expanded battery of cognitive tests with longitudinal follow-up and to identify the biological mechanisms underlying this relationship.
Collapse
Affiliation(s)
| | - Ryan K. Cvejkus
- Department of Epidemiology, University of Pittsburgh, Pi ttsburgh, PA, USA
| | - Joseph M. Zmuda
- Department of Epidemiology, University of Pittsburgh, Pi ttsburgh, PA, USA
| | - Allison L Kuipers
- Department of Epidemiology, University of Pittsburgh, Pi ttsburgh, PA, USA
| | | | - Akira Sekikawa
- Department of Epidemiology, University of Pittsburgh, Pi ttsburgh, PA, USA
| | - Victor Wheeler
- Tobago Health Studies Office, Scarborough, Tobago, Trinidad & Tobago
| | - Caterina Rosano
- Department of Epidemiology, University of Pittsburgh, Pi ttsburgh, PA, USA
| | - Iva Miljkovic
- Department of Epidemiology, University of Pittsburgh, Pi ttsburgh, PA, USA
| |
Collapse
|
38
|
Yu K, Siang Ng TK. Investigating Biological Pathways Underpinning the Longitudinal Association Between Loneliness and Cognitive Impairment. J Gerontol A Biol Sci Med Sci 2023; 78:1417-1426. [PMID: 36200979 PMCID: PMC11015400 DOI: 10.1093/gerona/glac213] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Loneliness precedes the onset of cognitive impairment (CI) in older adults. Although the mechanisms through which loneliness "gets under the skin" to influence the risk of developing CI have been conceptually proposed, they are rarely empirically examined. The Evolutionary Theory of Loneliness posits that loneliness as a stressor could cause dysregulations in multiple physiological systems. The current study investigated whether inflammatory, cardiovascular, and kidney biomarkers mediate the longitudinal association between loneliness and CI. METHODS Cross-lagged panel models were used to examine the hypothesized relationships, using 2006, 2010, and 2014 waves of data from the Health and Retirement Study (N = 7,037). Loneliness was measured with the 3-item UCLA loneliness scale. CI was assessed with the modified telephone interview for cognitive status. Biomarkers included HbA1C, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, C-reactive protein, and Cystatin C. Using a stepwise model-building approach, first, the model included only loneliness, CI, and biomarker variables; then, sociodemographic covariates were added; lastly, health status were controlled for. RESULTS In unadjusted and partially adjusted models, loneliness was associated with higher odds of worse cognitive status in an 8-year follow-up. Only HbA1C mediated the longitudinal association between loneliness and CI. However, after further controlling for health status, all associations became nonsignificant. CONCLUSIONS Examining a large number of participants and linking a limited number of biological markers with cognition and loneliness longitudinally, our empirical data did not support theoretical propositions, highlighting the critical importance of controlling for confounders in future studies examining longitudinal mediational relationships underlying loneliness and CI.
Collapse
Affiliation(s)
- Kexin Yu
- Suzanne Dworak-Peck School of Social Work, University of Southern California, Los Angeles, California, USA
- Edward R. Roybal Institute on Aging, University of Southern California, Los Angeles, California, USA
- NIA-Layton Aging and Alzheimer’s Disease Center, and Oregon Center for Aging and Technology, Department of Neurology, Oregon Health and Science University, Portland, Oregon, USA
| | - Ted Kheng Siang Ng
- Center for Innovation in Healthy and Resilient Aging (CIHRA), Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, Arizona, USA
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Center for Gait and Movement Analysis (CGMA), University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
39
|
Kim JS, Chen MH, Wang HE, Lu CL, Wang YP, Zhang B. Inflammatory Bowel Disease and Neurodegenerative Diseases. Gut Liver 2023; 17:495-504. [PMID: 36843420 PMCID: PMC10352055 DOI: 10.5009/gnl220523] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 02/28/2023] Open
Abstract
A growing body of evidence has demonstrated an intricate association between inflammatory bowel disease (IBD) and neurodegenerative conditions, expanding beyond previous foci of comorbidities between IBD and mood disorders. These new discoveries stem from an improved understanding of the gut-microbiome-brain axis: specifically, the ability of the intestinal microbiota to modulate inflammation and regulate neuromodulatory compounds. Clinical retrospective studies incorporating large sample sizes and population-based cohorts have demonstrated and confirmed the relevance of IBD and chronic neurodegeneration in clinical medicine. In this review, we expound upon the current knowledge on the gut-microbiome-brain axis, highlighting several plausible mechanisms linking IBD with neurodegeneration. We also summarize the known associations between IBD with Parkinson disease, Alzheimer disease, vascular dementia and ischemic stroke, and multiple sclerosis in a clinical context. Finally, we discuss the implications of an improved understanding of the gut-microbiome-brain axis in preventing, diagnosing, and managing neurodegeneration among IBD and non-IBD patients.
Collapse
Affiliation(s)
- Jin Sun Kim
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hohui E. Wang
- Department of Psychiatry, University of California San Francisco, San Francisco, CA, USA
| | - Ching-Liang Lu
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Endoscopy Center for Diagnosis and Treatment, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Po Wang
- Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Endoscopy Center for Diagnosis and Treatment, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Bing Zhang
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
40
|
Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther 2023; 8:267. [PMID: 37433768 PMCID: PMC10336149 DOI: 10.1038/s41392-023-01486-5] [Citation(s) in RCA: 263] [Impact Index Per Article: 131.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 07/13/2023] Open
Abstract
Studies in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and Amyotrophic lateral sclerosis, Huntington's disease, and so on, have suggested that inflammation is not only a result of neurodegeneration but also a crucial player in this process. Protein aggregates which are very common pathological phenomenon in neurodegeneration can induce neuroinflammation which further aggravates protein aggregation and neurodegeneration. Actually, inflammation even happens earlier than protein aggregation. Neuroinflammation induced by genetic variations in CNS cells or by peripheral immune cells may induce protein deposition in some susceptible population. Numerous signaling pathways and a range of CNS cells have been suggested to be involved in the pathogenesis of neurodegeneration, although they are still far from being completely understood. Due to the limited success of traditional treatment methods, blocking or enhancing inflammatory signaling pathways involved in neurodegeneration are considered to be promising strategies for the therapy of neurodegenerative diseases, and many of them have got exciting results in animal models or clinical trials. Some of them, although very few, have been approved by FDA for clinical usage. Here we comprehensively review the factors affecting neuroinflammation and the major inflammatory signaling pathways involved in the pathogenicity of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic lateral sclerosis. We also summarize the current strategies, both in animal models and in the clinic, for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Weifeng Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China
| | - Dan Xiao
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, P.R. China
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China.
| |
Collapse
|
41
|
Fulop T, Ramassamy C, Lévesque S, Frost EH, Laurent B, Lacombe G, Khalil A, Larbi A, Hirokawa K, Desroches M, Rodrigues S, Bourgade K, Cohen AA, Witkowski JM. Viruses - a major cause of amyloid deposition in the brain. Expert Rev Neurother 2023; 23:775-790. [PMID: 37551672 DOI: 10.1080/14737175.2023.2244162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
INTRODUCTION Clinically, Alzheimer's disease (AD) is a syndrome with a spectrum of various cognitive disorders. There is a complete dissociation between the pathology and the clinical presentation. Therefore, we need a disruptive new approach to be able to prevent and treat AD. AREAS COVERED In this review, the authors extensively discuss the evidence why the amyloid beta is not the pathological cause of AD which makes therefore the amyloid hypothesis not sustainable anymore. They review the experimental evidence underlying the role of microbes, especially that of viruses, as a trigger/cause for the production of amyloid beta leading to the establishment of a chronic neuroinflammation as the mediator manifesting decades later by AD as a clinical spectrum. In this context, the emergence and consequences of the infection/antimicrobial protection hypothesis are described. The epidemiological and clinical data supporting this hypothesis are also analyzed. EXPERT OPINION For decades, we have known that viruses are involved in the pathogenesis of AD. This discovery was ignored and discarded for a long time. Now we should accept this fact, which is not a hypothesis anymore, and stimulate the research community to come up with new ideas, new treatments, and new concepts.
Collapse
Affiliation(s)
- Tamas Fulop
- Research Center on Aging, Centre Intégré Universitaire de Santé Et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
- Department of Medicine, Division of Geriatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Simon Lévesque
- CIUSSS de l'Estrie - CHUS, Sherbrooke, QC, Canada
- Département de Microbiologie Et Infectiologie, Faculté de Médecine Et des Sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Eric H Frost
- Département de Microbiologie Et Infectiologie, Faculté de Médecine Et des Sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Benoit Laurent
- Research Center on Aging, Centre Intégré Universitaire de Santé Et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Guy Lacombe
- Research Center on Aging, Centre Intégré Universitaire de Santé Et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
- Department of Medicine, Division of Geriatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Abedelouahed Khalil
- Research Center on Aging, Centre Intégré Universitaire de Santé Et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
- Department of Medicine, Division of Geriatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Anis Larbi
- Department of Medicine, Division of Geriatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Katsuiku Hirokawa
- Department of Pathology, Institute of Health and Life Science, Tokyo Medical Dental University, Tokyo and Nito-Memory Nakanosogo Hospital, Tokyo, Japan
| | - Mathieu Desroches
- MathNeuro Team, Inria Sophia Antipolis Méditerranée, Biot, France
- Université Côte d'Azur, Nice, France
| | - Serafim Rodrigues
- Ikerbasque, BCAM, the Basque Foundation for Science and BCAM - The Basque Center for Applied Mathematics, Bilbao, Spain
| | - Karine Bourgade
- Research Center on Aging, Centre Intégré Universitaire de Santé Et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Alan A Cohen
- Department of Environmental Health Sciences, Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
42
|
Yang X, Ye T, He Y, Wen L, Cheng X. Qi-fu-yin attenuated cognitive disorders in 5xFAD mice of Alzheimer's disease animal model by regulating immunity. Front Neurol 2023; 14:1183764. [PMID: 37441611 PMCID: PMC10333586 DOI: 10.3389/fneur.2023.1183764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/01/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction Cognitive impairment is the main symptom of Alzheimer's disease (AD). Accumulating evidence implicate that immunity plays an important role in AD. Here, we investigated the effect of Qi-fu-yin (QFY) on cognitive impairment and cytokine secretion of 5xFAD mice. Methods We used 2.5-month-old 5xFAD transgenic mice for behavioral tests to observe the changes in cognitive function after QFY treatment. After the behavioral experiment, the whole brain, cortex and plasma of each mouse were collected for soluble Aβ analysis, immunohistochemical experiment and cytokine analysis. Results Here we found that the treatment of QFY ameliorated the ability of object recognition, passive avoidance responses and the ability of spatial learning and memory in 5xFAD mice. The deposits of β1 - 42 and Aβ1 - 40 were alleviated and the ration of Aβ1 - 42/Aβ1 - 40 was decrease in the plasma and brain of 5xFAD mice administrated with QFY. The administration of QFY promoted the secretion of anti-inflammatory cytokines, IL-5, IL-10 and G-CSF, and reduced the content of proinflammatory cytokines IFN-γ in plasma of 5xFAD mice. Notably, we found that the treatment of QFY decreased the concentration of CCL11 in the brain and plasma of 5xFAD mice. Conclusion This suggested that QFY improved cognition and reduced Aβ deposits in 5xFAD mice by regulating abnormal immunity in 5xFAD mice. QFY may be as a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Xiuzhao Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianyuan Ye
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yun He
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Wen
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology and Immunology Research, Department of Traditional Chinese Medicine, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaorui Cheng
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
43
|
Nidadavolu LS, Feger D, Chen D, Wu Y, Grodstein F, Gross AL, Bennett DA, Walston JD, Oh ES, Abadir PM. Associations between circulating cell-free mitochondrial DNA, inflammatory markers, and cognitive and physical outcomes in community dwelling older adults. Immun Ageing 2023; 20:24. [PMID: 37221566 PMCID: PMC10204157 DOI: 10.1186/s12979-023-00342-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: 02/22/2023] [Accepted: 04/21/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND Dementia and frailty are common age-related syndromes often linked to chronic inflammation. Identifying the biological factors and pathways that contribute to chronic inflammation is crucial for developing new therapeutic targets. Circulating cell-free mitochondrial DNA (ccf-mtDNA) has been proposed as an immune stimulator and potential predictor of mortality in acute illnesses. Dementia and frailty are both associated with mitochondrial dysfunction, impaired cellular energetics, and cell death. The size and abundance of ccf-mtDNA fragments may indicate the mechanism of cell death: long fragments typically result from necrosis, while short fragments arise from apoptosis. We hypothesize that increased levels of necrosis-associated long ccf-mtDNA fragments and inflammatory markers in serum are linked to declines in cognitive and physical function, as well as increased mortality risk. RESULTS Our study of 672 community-dwelling older adults revealed that inflammatory markers (C-Reactive Protein, soluble tumor necrosis factor alpha, tumor necrosis factor alpha receptor 1 [sTNFR1], and interleukin-6 [IL-6]) positively correlated with ccf-mtDNA levels in serum. Although cross-sectional analysis revealed no significant associations between short and long ccf-mtDNA fragments, longitudinal analysis demonstrated a connection between higher long ccf-mtDNA fragments (necrosis-associated) and worsening composite gait scores over time. Additionally, increased mortality risk was observed only in individuals with elevated sTNFR1 levels. CONCLUSION In a community dwelling cohort of older adults, there are cross-sectional and longitudinal associations between ccf-mtDNA and sTNFR1 with impaired physical and cognitive function and increased hazard of death. This work suggests a role for long ccf-mtDNA as a blood-based marker predictive of future physical decline.
Collapse
Affiliation(s)
- Lolita S Nidadavolu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Danielle Feger
- Johns Hopkins University Center on Aging and Health, Baltimore, MD, USA
| | - Diefei Chen
- Johns Hopkins University Center on Aging and Health, Baltimore, MD, USA
| | - Yuqiong Wu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Francine Grodstein
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Alden L Gross
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Johns Hopkins University Center on Aging and Health, Baltimore, MD, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jeremy D Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Esther S Oh
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Geriatric Medicine and Gerontology, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA.
| | - Peter M Abadir
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Geriatric Medicine and Gerontology, Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA.
| |
Collapse
|
44
|
Sommerlad A, Kivimäki M, Larson EB, Röhr S, Shirai K, Singh-Manoux A, Livingston G. Social participation and risk of developing dementia. NATURE AGING 2023; 3:532-545. [PMID: 37202513 DOI: 10.1038/s43587-023-00387-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/22/2023] [Indexed: 05/20/2023]
Abstract
The increasing number of people with dementia globally illustrates the urgent need to reduce dementia's scale and impact. Lifetime social participation may affect dementia risk by increasing cognitive reserve, and through brain maintenance by reducing stress and improving cerebrovascular health. It may therefore have important implications for individual behavior and public health policy aimed at reducing dementia burden. Observational study evidence indicates that greater social participation in midlife and late life is associated with 30-50% lower subsequent dementia risk, although some of this may not be causal. Social participation interventions have led to improved cognition but, partly due to short follow-up and small numbers of participants, no reduction in risk of dementia. We summarize the evidence linking social participation with dementia, discuss potential mechanisms by which social participation is likely to reduce and mitigate the impact of neuropathology in the brain, and consider the implications for future clinical and policy dementia prevention interventions.
Collapse
Affiliation(s)
- Andrew Sommerlad
- Division of Psychiatry, University College London, London, UK.
- Camden and Islington NHS Foundation Trust, London, UK.
| | - Mika Kivimäki
- Division of Psychiatry, University College London, London, UK
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Eric B Larson
- University of Washington Schools of Medicine and Public Health, Seattle, WA, USA
| | - Susanne Röhr
- School of Psychology, Massey University, Manawatu, New Zealand
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
- Institute of Social Medicine, Occupational Health and Public Health (ISAP), Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Kokoro Shirai
- Department of Social Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Archana Singh-Manoux
- Division of Psychiatry, University College London, London, UK
- Université Paris Cité, Inserm, U1153, Paris, France
| | - Gill Livingston
- Division of Psychiatry, University College London, London, UK
- Camden and Islington NHS Foundation Trust, London, UK
| |
Collapse
|
45
|
Duchaine CS, Brisson C, Diorio C, Talbot D, Maunsell E, Carmichael PH, Giguère Y, Gilbert-Ouimet M, Trudel X, Ndjaboué R, Vézina M, Milot A, Mâsse B, Dionne CE, Laurin D. Work-Related Psychosocial Factors and Global Cognitive Function: Are Telomere Length and Low-Grade Inflammation Potential Mediators of This Association? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4929. [PMID: 36981836 PMCID: PMC10049148 DOI: 10.3390/ijerph20064929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
The identification of modifiable factors that could maintain cognitive function is a public health priority. It is thought that some work-related psychosocial factors help developing cognitive reserve through high intellectual complexity. However, they also have well-known adverse health effects and are considered to be chronic psychosocial stressors. Indeed, these stressors could increase low-grade inflammation and promote oxidative stress associated with accelerated telomere shortening. Both low-grade inflammation and shorter telomeres have been associated with a cognitive decline. This study aimed to evaluate the total, direct, and indirect effects of work-related psychosocial factors on global cognitive function overall and by sex, through telomere length and an inflammatory index. A random sample of 2219 participants followed over 17 years was included in this study, with blood samples and data with cognitive function drawn from a longitudinal study of 9188 white-collar workers (51% female). Work-related psychosocial factors were evaluated according to the Demand-Control-Support and the Effort-Reward Imbalance (ERI) models. Global cognitive function was evaluated with the validated Montreal Cognitive Assessment (MoCA). Telomere length and inflammatory biomarkers were measured using standardised protocols. The direct and indirect effects were estimated using a novel mediation analysis method developed for multiple correlated mediators. Associations were observed between passive work or low job control, and shorter telomeres among females, and between low social support at work, ERI or iso-strain, and a higher inflammatory index among males. An association was observed with higher cognitive performance for longer telomeres, but not for the inflammatory index. Passive work overall, and low reward were associated with lower cognitive performance in males; whereas, high psychological demand in both males and females and high job strain in females were associated with a higher cognitive performance. However, none of these associations were mediated by telomere length or the inflammatory index. This study suggests that some work-related psychosocial factors could be associated with shorter telomeres and low-grade inflammation, but these associations do not explain the relationship between work-related psychosocial factors and global cognitive function. A better understanding of the biological pathways, by which these factors affect cognitive function, could guide future preventive strategies to maintain cognitive function and promote healthy aging.
Collapse
Affiliation(s)
- Caroline S. Duchaine
- Centre d’excellence sur le vieillissement de Québec (CEVQ), CIUSSS-Capitale Nationale, Québec, QC G1S 4L8, Canada
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
- VITAM, Centre de Recherche en santé Durable, Québec, QC G1S 4L8, Canada
- Institut sur le Vieillissement et la Participation Sociale des Aînés, Université Laval, Québec, QC G1S 4L8, Canada
| | - Chantal Brisson
- Centre d’excellence sur le vieillissement de Québec (CEVQ), CIUSSS-Capitale Nationale, Québec, QC G1S 4L8, Canada
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
- VITAM, Centre de Recherche en santé Durable, Québec, QC G1S 4L8, Canada
| | - Caroline Diorio
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
| | - Denis Talbot
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
| | - Elizabeth Maunsell
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
| | - Pierre-Hugues Carmichael
- Centre d’excellence sur le vieillissement de Québec (CEVQ), CIUSSS-Capitale Nationale, Québec, QC G1S 4L8, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
| | - Yves Giguère
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
| | - Mahée Gilbert-Ouimet
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
- Canada Research Chair in Sex and Gender in Occupational Health, Université du Québec à Rimouski, Campus de Lévis, Lévis, QC G6V 0A6, Canada
| | - Xavier Trudel
- Centre d’excellence sur le vieillissement de Québec (CEVQ), CIUSSS-Capitale Nationale, Québec, QC G1S 4L8, Canada
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
- VITAM, Centre de Recherche en santé Durable, Québec, QC G1S 4L8, Canada
| | - Ruth Ndjaboué
- School of Social Work, University of Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Michel Vézina
- Institut National de Santé Publique du Québec (INSPQ), Québec, QC G1V 5B3, Canada
| | - Alain Milot
- Centre d’excellence sur le vieillissement de Québec (CEVQ), CIUSSS-Capitale Nationale, Québec, QC G1S 4L8, Canada
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Benoît Mâsse
- École de Santé Publique de l’Université de Montréal, Montréal, QC H3N 1X9, Canada
| | - Clermont E. Dionne
- Centre d’excellence sur le vieillissement de Québec (CEVQ), CIUSSS-Capitale Nationale, Québec, QC G1S 4L8, Canada
- Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
- VITAM, Centre de Recherche en santé Durable, Québec, QC G1S 4L8, Canada
- Institut sur le Vieillissement et la Participation Sociale des Aînés, Université Laval, Québec, QC G1S 4L8, Canada
| | - Danielle Laurin
- Centre d’excellence sur le vieillissement de Québec (CEVQ), CIUSSS-Capitale Nationale, Québec, QC G1S 4L8, Canada
- Centre de Recherche du CHU de Québec—Université Laval, Québec, QC G1S 4L8, Canada
- VITAM, Centre de Recherche en santé Durable, Québec, QC G1S 4L8, Canada
- Institut sur le Vieillissement et la Participation Sociale des Aînés, Université Laval, Québec, QC G1S 4L8, Canada
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
46
|
De Vlieger L, Vandenbroucke RE, Van Hoecke L. Recent insights into viral infections as a trigger and accelerator in alzheimer's disease. Drug Discov Today 2022; 27:103340. [PMID: 35987492 PMCID: PMC9385395 DOI: 10.1016/j.drudis.2022.103340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/08/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder for which only symptomatic medication is available, except for the recently FDA-approved aducanumab. This lack of effective treatment urges us to investigate alternative paths that might contribute to disease development. In light of the recent SARS-CoV-2 pandemic and the disturbing neurological complications seen in some patients, it is desirable to (re)investigate the viability of the viral infection theory claiming that a microbe could affect AD initiation and/or progression. Here, we review the most important evidence for this theory with a special focus on two viruses, namely HSV-1 and SARS-CoV-2. Moreover, we discuss the possible involvement of extracellular vesicles (EVs). This overview will contribute to a more rational approach of potential treatment strategies for AD patients.
Collapse
Affiliation(s)
- Lize De Vlieger
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Lien Van Hoecke
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
47
|
Morrison HW, White MM, Rothers JL, Taylor-Piliae RE. Examining the Associations between Post-Stroke Cognitive Function and Common Comorbid Conditions among Stroke Survivors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13445. [PMID: 36294026 PMCID: PMC9603222 DOI: 10.3390/ijerph192013445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/03/2022] [Accepted: 10/15/2022] [Indexed: 06/16/2023]
Abstract
A considerable complication for stroke survivors is the subsequent development of cognitive decline or dementia. In this study, the relationship between the inflammation-centered comorbidity burden on post-stroke cognitive function among community-dwelling stroke survivors capable of independent living was examined. Data for this secondary analysis were collected from stroke survivors (n = 97) participating in a randomized clinical trial. Participants provided baseline responses, regarding cognitive function (mini-mental status exam, MMSE; Montreal cognitive assessment, MoCA), history of stroke comorbid conditions, and the Stroke Prognosis Instrument-II (SPI-II), an index of stroke comorbidity and recurrent stroke risk within the next two years. Relationships and differences between groups were tested for significance using Spearman's correlation, Kruskal-Wallis, or Mann-Whitney U tests. Most stroke survivors (69%) had multiple comorbidities. Total SPI-II scores were negatively correlated to both MoCA and MMSE scores (r = -0.25, p = 0.01; r = -0.22, p = 0.03, respectively), and differences in MoCA scores among SPI-II risk groups (low, medium, high) were evident (p = 0.05). In contrast, there were no differences in MoCA or MMSE scores when comorbid conditions were examined individually. Lastly, no gender differences were evident in cognitive assessments. Our data support the premise that comorbidity's burden impacts post-stroke cognitive decline, more than a single comorbid condition. Inflammation may be an important component of this comorbidity burden. Future studies that operationalize this concept will better illuminate the complex phenomenon of post-stroke cognitive decline for improved clinical rehabilitation modalities.
Collapse
Affiliation(s)
| | - Melissa M. White
- El Paso Veteran’s Administration Healthcare System, El Paso, TX 79930, USA
| | - Janet L. Rothers
- BIO5 Institute Statistics Consulting Lab, The University of Arizona, Tucson, AZ 85721, USA
| | | |
Collapse
|
48
|
Zhang Y, Song H, Wang J, Xi X, Cefalo P, Wood LJ, Luo X, Wang QM. Multiplex array analysis of serum cytokines offers minimal predictive value for cognitive function in the subacute phase after stroke. Front Neurol 2022; 13:886018. [PMID: 36330425 PMCID: PMC9622930 DOI: 10.3389/fneur.2022.886018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/29/2022] [Indexed: 10/28/2023] Open
Abstract
OBJECTIVE The effects of inflammation on post-stroke cognitive function are still unclear. This study investigated the correlation between the Th17-related cytokines in peripheral blood and post-stroke cognitive function after ischemic stroke in the subacute phase. DESIGN A retrospective cohort study. SETTING Academic acute inpatient rehabilitation facility. PARTICIPANTS One hundred and fourteen patients with first ischemic stroke were categorized as the poor cognitive recovery group (n = 58) or good cognitive recovery group (n = 56) based on their cognitive MRFS efficiency. INTERVENTIONS All subjects received routine physical, occupational, and speech-language pathology therapy. MAIN OUTCOME MEASURES Serum cytokines/chemokine (IL-1 β, IL-2, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12p70, IL-13, IL-15, IL-17A, IL-17E, IL-17F, IL-21, IL-22, IL-23, IL-27, IL-28A, IL-31, IL-33, GM-CSF, IFN-γ, MIP-3 α, TNF-α, and TNF-β) levels were measured in duplicate using Human Th17 magnetic bead panel and multiplex array analysis (Luminex-200 system). The primary functional outcome was a gain in functional independence measure (FIM) cognitive subscore at discharge. The secondary outcome measures were FIM total score at discharge, length of stay in the hospital, and discharge destination. Cognitive Montebello Rehabilitation Factor Score (MRFS) and cognitive MRFS efficiency were calculated. Demographic and clinical characteristics were obtained from the medical record. RESULTS The good cognitive recovery group had an interesting trend of higher IL-13 than the poor cognitive recovery group (good cognitive recovery group 257.82 ± 268.76 vs. poor cognitive recovery group 191.67 ± 201.82, p = 0.049, unit: pg/ml). However, Pearson's correlation analysis showed no significant correlation between cytokine levels and gain of cognition, cognitive MRFS, or cognitive MRFS efficiency. Receiver operating characteristic (ROC) analysis of cytokines also suggested a low accuracy of prediction as a predictor for post-stroke cognitive recovery improvement. CONCLUSION Our preliminary findings suggested that the level of serum cytokines had minimal predictive value for the recovery of cognitive function during the subacute inpatient rehabilitation after stroke.
Collapse
Affiliation(s)
- Yuling Zhang
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, The Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Haixin Song
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, The Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
- Rehabilitation Department, Sir Run Run Show Hospital, Hangzhou, China
| | - Jun Wang
- School of Medicine, Shenzhen University, Shenzhen, China
| | - Xiao Xi
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, The Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
- Department of Rehabilitation Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Philip Cefalo
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, The Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
| | - Lisa J. Wood
- William F. Connell School of Nursing at Boston College, Boston, MA, United States
| | - Xun Luo
- School of Medicine, Shenzhen University, Shenzhen, China
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, China
| | - Qing Mei Wang
- Stroke Biological Recovery Laboratory, Spaulding Rehabilitation Hospital, The Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, The Teaching Affiliate of Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
49
|
Ebright B, Assante I, Poblete RA, Wang S, Duro MV, Bennett DA, Arvanitakis Z, Louie SG, Yassine HN. Eicosanoid lipidome activation in post-mortem brain tissues of individuals with APOE4 and Alzheimer's dementia. Alzheimers Res Ther 2022; 14:152. [PMID: 36217192 PMCID: PMC9552454 DOI: 10.1186/s13195-022-01084-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Chronic neuroinflammation is one of the hallmarks of late-onset Alzheimer's disease (AD) dementia pathogenesis. Carrying the apolipoprotein ε4 (APOE4) allele has been associated with an accentuated response to brain inflammation and increases the risk of AD dementia progression. Among inflammation signaling pathways, aberrant eicosanoid activation plays a prominent role in neurodegeneration. METHODS Using brains from the Religious Order Study (ROS), this study compared measures of brain eicosanoid lipidome in older persons with AD dementia to age-matched controls with no cognitive impairment (NCI), stratified by APOE genotype. RESULTS Lipidomic analysis of the dorsolateral prefrontal cortex demonstrated lower levels of omega-3 fatty acids eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and DHA-derived neuroprotectin D1 (NPD-1) in persons with AD dementia, all of which associated with lower measures of cognitive function. A significant interaction was observed between carrying the APOE4 allele and higher levels of both pro-inflammatory lipids and pro-resolving eicosanoid lipids on measures of cognitive performance and on neuritic plaque burden. Furthermore, analysis of lipid metabolism pathways implicated activation of calcium-dependent phospholipase A2 (cPLA2), 5-lipoxygenase (5-LOX), and soluble epoxide hydrolase (sEH) enzymes. CONCLUSION These findings implicate activation of the eicosanoid lipidome in the chronic unresolved state of inflammation in AD dementia, which is increased in carriers of the APOE4 allele, and identify potential therapeutic targets for resolving this chronic inflammatory state.
Collapse
Affiliation(s)
- Brandon Ebright
- School of Pharmacy, University of Southern California, Los Angeles, USA
| | - Isaac Assante
- School of Pharmacy, University of Southern California, Los Angeles, USA
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Roy A Poblete
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Shaowei Wang
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Marlon V Duro
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Zoe Arvanitakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Stan G Louie
- School of Pharmacy, University of Southern California, Los Angeles, USA.
- Keck School of Medicine, University of Southern California, Los Angeles, USA.
| | - Hussein N Yassine
- Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
50
|
Tyler SEB, Tyler LDK. Therapeutic roles of plants for 15 hypothesised causal bases of Alzheimer's disease. NATURAL PRODUCTS AND BIOPROSPECTING 2022; 12:34. [PMID: 35996065 PMCID: PMC9395556 DOI: 10.1007/s13659-022-00354-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/15/2022] [Indexed: 05/26/2023]
Abstract
Alzheimer's disease (AD) is progressive and ultimately fatal, with current drugs failing to reverse and cure it. This study aimed to find plant species which may provide therapeutic bioactivities targeted to causal agents proposed to be driving AD. A novel toolkit methodology was employed, whereby clinical symptoms were translated into categories recognized in ethnomedicine. These categories were applied to find plant species with therapeutic effects, mined from ethnomedical surveys. Survey locations were mapped to assess how this data is at risk. Bioactivities were found of therapeutic relevance to 15 hypothesised causal bases for AD. 107 species with an ethnological report of memory improvement demonstrated therapeutic activity for all these 15 causal bases. The majority of the surveys were found to reside within biodiversity hotspots (centres of high biodiversity under threat), with loss of traditional knowledge the most common threat. Our findings suggest that the documented plants provide a large resource of AD therapeutic potential. In demonstrating bioactivities targeted to these causal bases, such plants may have the capacity to reduce or reverse AD, with promise as drug leads to target multiple AD hallmarks. However, there is a need to preserve ethnomedical knowledge, and the habitats on which this knowledge depends.
Collapse
Affiliation(s)
| | - Luke D K Tyler
- School of Natural Sciences, Bangor University, Gwynedd, UK
| |
Collapse
|