1
|
Mirzaei H, Patrova J, Mannheimer B, Lindh JD, Falhammar H. Prevalence and Incidence of Dementia in Patients With Non-Overtly Functional Adrenal Tumours. Clin Endocrinol (Oxf) 2024. [PMID: 39722568 DOI: 10.1111/cen.15186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE To investigate the prevalence and incidence of dementia in patients with non-overtly functional adrenal tumours (NOFATs). DESIGN A national retrospective register-based study was conducted on patients diagnosed with NOFAT and controls diagnosed between 2005 and 2019, in Sweden. Individuals diagnosed with overt adrenal hormone excess or previous malignancies were excluded. Sensitivity analyses were performed in subgroups with a combination of gallbladder/biliary tract/pancreatic disease. and acute appendicitis, as well as 3- and 12-months of malignancy-free survival. MEASUREMENTS Prevalence and incidence of dementia. The secondary outcomes were Alzheimer's disease and vascular dementia. RESULTS Among 20,390 cases, 12,120 (59.4%) were women, and the median (IQR) age was 66 (57-73) years. Among the 125,392 controls, 69,994 (55.8%) were women and the median (IQR) age was 66 (57-73) years. Patients with NOFATs had a lower prevalence of dementia compared to controls (odds ratio [OR] 0.58, 95% CI 0.50-0.68, adjusted OR [aOR] 0.47, 95% CI 0.40-0.56). During the follow-up period (median 4.9 years, IQR 2.2-8.2), incidence of dementia was similar in NOFATs and controls (hazard ratio [HR] 1.05, 95% CI 0.97-1.15, adjusted HR [aHR] 1.06, 95% CI 0.97-1.15). Similar results obtained for Alzheimer's dementia (aOR 0.44, 95% CI 0.34-0.57; aHR 0.94, 95% CI 0.80-1.10) and vascular dementia (OR 0.71, 95% CI 0.52-0.94, aOR 0.48, 95% CI 0.35-0.64; HR 1.29, 95% CI 1.08-1.53, aHR 1.13, 95% CI 0.95-1.35) as well as in the sensitivity analyses. Adrenalectomy did not change the results. CONCLUSION NOFAT was not associated with an increased risk of dementia.
Collapse
Affiliation(s)
- Hadis Mirzaei
- Department of Endocrinology, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science ad Education, Södersjukhuset, Karolinska Institute, Stockholm, Sweden
| | - Jekaterina Patrova
- Department of Endocrinology, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science ad Education, Södersjukhuset, Karolinska Institute, Stockholm, Sweden
| | - Buster Mannheimer
- Department of Endocrinology, Södersjukhuset, Stockholm, Sweden
- Department of Clinical Science ad Education, Södersjukhuset, Karolinska Institute, Stockholm, Sweden
| | - Jonatan D Lindh
- Laboratory Medicine, Division of Clinical Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
2
|
Wang T, Li X, Jia Y, Zhang Y, Meng D. Effects of cortisol on cognitive and emotional disorders after stroke: A scoping review. Heliyon 2024; 10:e40278. [PMID: 39634426 PMCID: PMC11616494 DOI: 10.1016/j.heliyon.2024.e40278] [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: 03/14/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Objectives Stroke-induced cognitive and mood disorders are closely related to glucocorticoids released during hypothalamic-pituitary-adrenal (HPA) axis activation. There are many studies on the relationship between cortisol levels and post-stroke cognitive impairment (PSCI) and post-stroke depression (PSD). This paper provides a scoping review of these studies to clarify the effect of cortisol on PSCI and PSD, thereby providing a theoretical basis for clinical diagnosis and treatment. Materials and methods We searched for literature published up to October 2023 on the association of cortisol with post-stroke cognitive and emotional disorders in the PubMed, Web of Science, Cochrane Library, CNKI and Wanfang databases. Relevant papers were identified and the effects of cortisol on cognitive and emotional disorders after stroke were analyzed by literature induction. Results Eighteen papers were included, including cross-sectional studies and cohort studies. The subjects suffered ischemic stroke or hemorrhagic stroke. Cortisol levels were measured from samples of blood, saliva or hair. Most patients showed increased basal cortisol levels and changes in cortisol circadian rhythms. Most studies report that patients with high cortisol levels on admission (acute phase of stroke) are more likely to experience cognitive decline and depression later in life. Conclusions Admission cortisol level may be a promising biomarker for predicting cognitive and emotional prognosis after stroke.
Collapse
Affiliation(s)
- Tiantian Wang
- Rehabilitation Medical Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Xuan Li
- Rehabilitation Medical Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Yuanyuan Jia
- Rehabilitation Medical Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Yuyao Zhang
- Rehabilitation Medical Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Dianhuai Meng
- Rehabilitation Medical Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
3
|
El Abiad E, Al-Kuwari A, Al-Aani U, Al Jaidah Y, Chaari A. Navigating the Alzheimer's Biomarker Landscape: A Comprehensive Analysis of Fluid-Based Diagnostics. Cells 2024; 13:1901. [PMID: 39594648 PMCID: PMC11593284 DOI: 10.3390/cells13221901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) affects a significant portion of the aging population, presenting a serious challenge due to the limited availability of effective therapies during its progression. The disease advances rapidly, underscoring the need for early diagnosis and the application of preventative measures. Current diagnostic methods for AD are often expensive and invasive, restricting access for the general public. One potential solution is the use of biomarkers, which can facilitate early detection and treatment through objective, non-invasive, and cost-effective evaluations of AD. This review critically investigates the function and role of biofluid biomarkers in detecting AD, with a specific focus on cerebrospinal fluid (CSF), blood-based, and saliva biomarkers. RESULTS CSF biomarkers have demonstrated potential for accurate diagnosis and valuable prognostic insights, while blood biomarkers offer a minimally invasive and cost-effective approach for diagnosing cognitive issues. However, while current biomarkers for AD show significant potential, none have yet achieved the precision needed to replace expensive PET scans and CSF assays. The lack of a single accurate biomarker underscores the need for further research to identify novel or combined biomarkers to enhance the clinical efficacy of existing diagnostic tests. In this context, artificial intelligence (AI) and deep-learning (DL) tools present promising avenues for improving biomarker analysis and interpretation, enabling more precise and timely diagnoses. CONCLUSIONS Further research is essential to confirm the utility of all AD biomarkers in clinical settings. Combining biomarker data with AI tools offers a promising path toward revolutionizing the personalized characterization and early diagnosis of AD symptoms.
Collapse
Affiliation(s)
| | | | | | | | - Ali Chaari
- Weill Cornell Medicine–Qatar, Qatar Foundation, Education City, Doha P.O. Box 24144, Qatar; (E.E.A.); (A.A.-K.); (U.A.-A.); (Y.A.J.)
| |
Collapse
|
4
|
Farnum Z, Mani R, Bindoff A, Wilson R, Fiotakis A, Stephens J, Cho E, Mackay-Sim A, Sinclair D. Convergent effects of synthetic glucocorticoid dexamethasone and amyloid beta in human olfactory neurosphere-derived cells. J Neurochem 2024. [PMID: 39556451 DOI: 10.1111/jnc.16263] [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: 06/18/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024]
Abstract
Stressful life events and glucocorticoid (stress) hormones appear to increase the risk of Alzheimer's disease and hasten its progression, but the reasons for this remain unclear. One potential explanation is that when amyloid β (Aβ) pathology is accumulating in the preclinical disease stage, glucocorticoid receptor signalling during stressful events exacerbates cellular dysfunction caused by Aβ. Alternatively, Aβ may disrupt glucocorticoid receptor signalling. To explore these possibilities, we investigated whether the synthetic glucocorticoid dexamethasone and Aβ have overlapping effects on the cellular proteome and whether Aβ influences canonical glucocorticoid receptor function. Human olfactory neurosphere-derived (ONS) cells, collected from the olfactory mucosa of six adult donors, were treated with soluble Aβ40 or Aβ42 followed by dexamethasone. Proteins were quantified by mass spectrometry. After 32 h treatment, Aβ40 and Aβ42 both induced profound changes in innate immunity-related proteins. After 72 h, Aβ42 formed widespread aggregates and induced few proteomic changes, whereas Aβ40 remained soluble and altered expression of mitochondrial and innate immunity-related proteins. ONS cells revealed overlapping impacts of Aβ40 and dexamethasone, with 23 proteins altered by both treatments. For 16 proteins (including eight mitochondrial proteins) dexamethasone counteracted the effects of Aβ40. For example, caspase 4 and methylmalonate-semialdehyde dehydrogenase were increased by Aβ40 and decreased by dexamethasone. Consistent with this finding, Aβ40 increased, but dexamethasone decreased, ONS cell proliferation. For seven proteins, including superoxide dismutase [Mn] mitochondrial, dexamethasone exacerbated the effects of Aβ40. For some proteins, including complement C3, the effects of dexamethasone differed depending on whether Aβ40 was present or absent. Neither Aβ species influenced glucocorticoid receptor nuclear translocation. Overall, this study revealed that glucocorticoid receptor signalling modifies the intracellular effects of Aß40, counteracting some effects and exacerbating others. It suggests that cellular mechanisms through which glucocorticoid receptor signalling influences Alzheimer's disease risk/progression are complex and determined by the balance of beneficial and detrimental glucocorticoid effects.
Collapse
Affiliation(s)
- Zane Farnum
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Radhika Mani
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Aidan Bindoff
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Richard Wilson
- Central Science Laboratory, University of Tasmania, Hobart, Tasmania, Australia
| | - Adoni Fiotakis
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Jessica Stephens
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Ellie Cho
- Biological Optical Microscopy Platform, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alan Mackay-Sim
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - Duncan Sinclair
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
5
|
Sibilia F, Sheikh-Bahaei N, Mack WJ, Barisano G, Choupan J. Neuroinflammation modifies the relationship between stress and perivascular spaces in an elderly population with different levels of cognitive impairment. Front Cell Neurosci 2024; 18:1480405. [PMID: 39610697 PMCID: PMC11603360 DOI: 10.3389/fncel.2024.1480405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024] Open
Abstract
Background Perivascular spaces (PVS) are fluid-filled spaces surrounding the brain parenchymal vasculature. Literature suggests that PVS may play a significant role in aging and neurological disorders, including Alzheimer's disease (AD). The aim of this study is to investigate whether the relationship between MRI-visible PVS and stress is influenced by neuroinflammation in an elderly population with different levels of cognitive impairment. Methods Using brain MRI scans acquired at 1.5 T, PVS were quantified in a cohort of 461 individuals, consisting of cognitively healthy controls (n = 48), people with mild cognitive impairment (MCI, n = 322) and Alzheimer's disease (AD, n = 91). PVS volume fraction was calculated in the basal ganglia and centrum semiovale using a semi-automated segmentation approach. Stress was quantified with levels of salivary cortisol. Inflammatory biomarkers measured from plasma included cytokines, matrix metalloproteinases and C-reactive protein. General linear models were used to test the relationship between PVS and cortisol, when interacting with inflammatory markers. This was done on the whole cohort and within each clinical cognitive group. Results In the centrum semiovale, higher inflammation levels reduced the relationship of cortisol with PVS. In basal ganglia, higher levels of C-reactive protein reduced the negative relationship of cortisol with PVS. All analyses were accounted for age, sex, body mass index (BMI) and total hippocampal volume. There was a significant interaction effect between cortisol and C-reactive protein on PVS volume fraction in the MCI group. Discussion These findings suggest an influence of neuroinflammation on the PVS structure in Alzheimer's disease spectrum, and offer insight for better understanding physiological processes of cognitive impairment onset.
Collapse
Affiliation(s)
- Francesca Sibilia
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Nasim Sheikh-Bahaei
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Wendy J. Mack
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Giuseppe Barisano
- Department of Neurosurgery, Stanford University, Stanford, CA, United States
| | - Jeiran Choupan
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- NeuroScope Inc,Scarsdale, NY, United States
| |
Collapse
|
6
|
Daniilidou M, Holleman J, Hagman G, Kåreholt I, Aspö M, Brinkmalm A, Zetterberg H, Blennow K, Solomon A, Kivipelto M, Sindi S, Matton A. Neuroinflammation, cerebrovascular dysfunction and diurnal cortisol biomarkers in a memory clinic cohort: Findings from the Co-STAR study. Transl Psychiatry 2024; 14:364. [PMID: 39251589 PMCID: PMC11385239 DOI: 10.1038/s41398-024-03072-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024] Open
Abstract
Cortisol dysregulation, neuroinflammation, and cerebrovascular dysfunction are biological processes that have been separately shown to be affected in Alzheimer's disease (AD). Here, we aimed to identify biomarker signatures reflecting these pathways in 108 memory clinic patients with subjective cognitive decline (SCD, N = 40), mild cognitive impairment (MCI, N = 39), and AD (N = 29). Participants were from the well-characterized Cortisol and Stress in Alzheimer's Disease (Co-STAR) cohort, recruited at Karolinska University Hospital. Salivary diurnal cortisol measures and 41 CSF proteins were analyzed. Principal component analysis was applied to identify combined biosignatures related to AD pathology, synaptic loss, and neuropsychological assessments, in linear regressions adjusted for confounders, such as age, sex, education and diagnosis. We found increased CSF levels of C-reactive protein (CRP), interferon γ-inducible protein (IP-10), thymus and activation-regulated chemokine (TARC), intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) in MCI patients. Further, markers of cortisol dysregulation (flattened salivary cortisol awakening response and flattened cortisol slope) correlated with increased levels of placental growth factor (PlGF), IP-10, and chitinase 3-like 1 (YKL-40) in the total cohort. A biosignature composed of cortisol awakening response, cortisol slope, and CSF IL-6 was downregulated in AD patients. Moreover, biomarker signatures reflecting overlapping pathophysiological processes of neuroinflammation and vascular injury were associated with AD pathology, synaptic loss, and worsened processing speed. Our findings suggest an early dysregulation of immune and cerebrovascular processes during the MCI stage and provide insights into the interrelationship of chronic stress and neuroinflammation in AD.
Collapse
Affiliation(s)
- Makrina Daniilidou
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Jasper Holleman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Göran Hagman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Ingemar Kåreholt
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Institute of Gerontology, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Malin Aspö
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Alina Solomon
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, Kuopio, Finland
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Shireen Sindi
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Anna Matton
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
7
|
Krolick KN, Cao J, Gulla EM, Bhardwaj M, Marshall SJ, Zhou EY, Kiss AJ, Choueiry F, Zhu J, Shi H. Subregion-specific transcriptomic profiling of rat brain reveals sex-distinct gene expression impacted by adolescent stress. Neuroscience 2024; 553:19-39. [PMID: 38977070 PMCID: PMC11444371 DOI: 10.1016/j.neuroscience.2024.07.002] [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: 01/18/2024] [Revised: 05/14/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Stress during adolescence clearly impacts brain development and function. Sex differences in adolescent stress-induced or exacerbated emotional and metabolic vulnerabilities could be due to sex-distinct gene expression in hypothalamic, limbic, and prefrontal brain regions. However, adolescent stress-induced whole-genome expression changes in key subregions of these brain regions were unclear. In this study, female and male adolescent Sprague Dawley rats received one-hour restraint stress daily from postnatal day (PD) 32 to PD44. Corticosterone levels, body weights, food intake, body composition, and circulating adiposity and sex hormones were measured. On PD44, brain and blood samples were collected. Using RNA-sequencing, sex-specific differences in stress-induced differentially expressed (DE) genes were identified in subregions of the hypothalamus, limbic system, and prefrontal cortex. Canonical pathways reflected well-known sex-distinct maladies and diseases, substantiating the therapeutic potential of the DE genes found in the current study. Thus, we proposed specific sex distinct, adolescent stress-induced transcriptional changes found in the current study as examples of the molecular bases for sex differences witnessed in stress induced or exacerbated emotional and metabolic disorders. Future behavioral studies and single-cell studies are warranted to test the implications of the DE genes identified in this study in sex-distinct stress-induced susceptibilities.
Collapse
Affiliation(s)
| | - Jingyi Cao
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Evelyn M Gulla
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Meeta Bhardwaj
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | | | - Ethan Y Zhou
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Andor J Kiss
- Center for Bioinformatics & Functional Genomics, Miami University, Oxford, OH 45056, USA.
| | - Fouad Choueiry
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA.
| | - Jiangjiang Zhu
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
8
|
Chen Y, Al-Nusaif M, Li S, Tan X, Yang H, Cai H, Le W. Progress on early diagnosing Alzheimer's disease. Front Med 2024; 18:446-464. [PMID: 38769282 PMCID: PMC11391414 DOI: 10.1007/s11684-023-1047-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/15/2023] [Indexed: 05/22/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects both cognition and non-cognition functions. The disease follows a continuum, starting with preclinical stages, progressing to mild cognitive and behavioral impairment, ultimately leading to dementia. Early detection of AD is crucial for better diagnosis and more effective treatment. However, the current AD diagnostic tests of biomarkers using cerebrospinal fluid and/or brain imaging are invasive or expensive, and mostly are still not able to detect early disease state. Consequently, there is an urgent need to develop new diagnostic techniques with higher sensitivity and specificity during the preclinical stages of AD. Various non-cognitive manifestations, including behavioral abnormalities, sleep disturbances, sensory dysfunctions, and physical changes, have been observed in the preclinical AD stage before occurrence of notable cognitive decline. Recent research advances have identified several biofluid biomarkers as early indicators of AD. This review focuses on these non-cognitive changes and newly discovered biomarkers in AD, specifically addressing the preclinical stages of the disease. Furthermore, it is of importance to explore the potential for developing a predictive system or network to forecast disease onset and progression at the early stage of AD.
Collapse
Affiliation(s)
- Yixin Chen
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Murad Al-Nusaif
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Xiang Tan
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huijia Yang
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China.
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China.
| |
Collapse
|
9
|
Sinclair D, Canty AJ, Ziebell JM, Woodhouse A, Collins JM, Perry S, Roccati E, Kuruvilla M, Leung J, Atkinson R, Vickers JC, Cook AL, King AE. Experimental laboratory models as tools for understanding modifiable dementia risk. Alzheimers Dement 2024; 20:4260-4289. [PMID: 38687209 PMCID: PMC11180874 DOI: 10.1002/alz.13834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/29/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024]
Abstract
Experimental laboratory research has an important role to play in dementia prevention. Mechanisms underlying modifiable risk factors for dementia are promising targets for dementia prevention but are difficult to investigate in human populations due to technological constraints and confounds. Therefore, controlled laboratory experiments in models such as transgenic rodents, invertebrates and in vitro cultured cells are increasingly used to investigate dementia risk factors and test strategies which target them to prevent dementia. This review provides an overview of experimental research into 15 established and putative modifiable dementia risk factors: less early-life education, hearing loss, depression, social isolation, life stress, hypertension, obesity, diabetes, physical inactivity, heavy alcohol use, smoking, air pollution, anesthetic exposure, traumatic brain injury, and disordered sleep. It explores how experimental models have been, and can be, used to address questions about modifiable dementia risk and prevention that cannot readily be addressed in human studies. HIGHLIGHTS: Modifiable dementia risk factors are promising targets for dementia prevention. Interrogation of mechanisms underlying dementia risk is difficult in human populations. Studies using diverse experimental models are revealing modifiable dementia risk mechanisms. We review experimental research into 15 modifiable dementia risk factors. Laboratory science can contribute uniquely to dementia prevention.
Collapse
Affiliation(s)
- Duncan Sinclair
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Alison J. Canty
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
- Global Brain Health Institute, Trinity CollegeDublinIreland
| | - Jenna M. Ziebell
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Jessica M. Collins
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Sharn Perry
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Eddy Roccati
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Maneesh Kuruvilla
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Jacqueline Leung
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Rachel Atkinson
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - James C. Vickers
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Anthony L. Cook
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| | - Anna E. King
- Wicking Dementia Research and Education Centre, University of TasmaniaHobartTasmaniaAustralia
| |
Collapse
|
10
|
Dos Santos HM, Bertollo AG, Mingoti MED, Grolli RE, Kreuz KM, Ignácio ZM. Dementia and depression: Biological connections with amyloid β protein. Basic Clin Pharmacol Toxicol 2024; 134:563-573. [PMID: 38459754 DOI: 10.1111/bcpt.13996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/10/2024]
Abstract
Dementia is an umbrella term for a broad group of age-associated neurodegenerative diseases. It is estimated that dementia affects 50 million people worldwide and that Alzheimer's disease (AD) is responsible for up to 75% of cases. Small extracellular senile plaques composed of filamentous aggregates of amyloid β (Aβ) protein tend to bind to neuronal receptors, affecting cholinergic, serotonergic, dopaminergic and noradrenergic neurotransmission, leading to neuroinflammation, among other pathophysiologic processes and subsequent neuronal death, followed by dementia. The amyloid cascade hypothesis points to a pathological process in the cleavage of the amyloid precursor protein (APP), resulting in pathological Aβ. There is a close relationship between the pathologies that lead to dementia and depression. It is estimated that depression is prevalent in up to 90% of individuals diagnosed with Parkinson's disease, with varying severity, and in 20 to 30% of cases of Alzheimer's disease. The hypothalamic pituitary adrenal (HPA) axis is the great intermediary between the pathophysiological mechanisms in neurodegenerative diseases and depression. This review discusses the role of Aβ protein in the pathophysiological mechanisms of dementia and depression, considering the HPA axis, neuroinflammation, oxidative stress, signalling pathways and neurotransmission.
Collapse
Affiliation(s)
- Helamã Moraes Dos Santos
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Amanda Gollo Bertollo
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Maiqueli Eduarda Dama Mingoti
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Roberta Eduarda Grolli
- Laboratory for research into care, patient safety, and technological innovation in nursing and health, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Kelli Maria Kreuz
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Zuleide Maria Ignácio
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| |
Collapse
|
11
|
Lopez M, Siedl A, Phillips KA. Cortisol levels across the lifespan in common marmosets (Callithrix jacchus). Am J Primatol 2024; 86:e23597. [PMID: 38239052 PMCID: PMC10959686 DOI: 10.1002/ajp.23597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/13/2023] [Accepted: 12/28/2023] [Indexed: 02/13/2024]
Abstract
Human aging is associated with senescence of the hypothalamic-pituitary-adrenal (HPA) axis, leading to progressive dysregulation characterized by increased cortisol exposure. This key hormone is implicated in the pathogenesis of many age-related diseases. Common marmosets (Callithrix jacchus) display a wide spectrum of naturally occurring age-related pathologies that compare similarly to humans and are increasingly used as translational models of aging and age-related disease. Whether the marmoset HPA axis also shows senescence with increasing age is unknown. We analyzed hair cortisol concentration (HCC) across the lifespan of 50 captive common marmosets, ranging in age from approximately 2 months-14.5 years, via a cross-sectional design. Samples were processed and analyzed for cortisol using enzyme immunoassay. HCC ranged from 1416 to 15,343 pg/mg and was negatively correlated with age. We found significant main effects of age group (infant, adolescent, adult, aged, very aged) and sex on HCC, and no interaction effects. Infants had significantly higher levels of HCC compared with all other age groups. Females had higher HCC than males. There was no interaction between age and sex. These results suggest marmosets do not show dysregulation of the HPA axis with increasing age, as measured via HCC.
Collapse
Affiliation(s)
- Matthew Lopez
- Department of Psychology, Trinity University, 607 Kings Court, San Antonio Texas, USA
| | - Amaya Siedl
- Department of Psychology, Trinity University, 607 Kings Court, San Antonio Texas, USA
| | - Kimberley A. Phillips
- Department of Psychology, Trinity University, 607 Kings Court, San Antonio Texas, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas USA
| |
Collapse
|
12
|
Garbarino VR, Palavicini JP, Melendez J, Barthelemy N, He Y, Kautz TF, Lopez-Cruzan M, Mathews JJ, Xu P, Zhan B, Saliba A, Ragi N, Sharma K, Craft S, Petersen RC, Espindola-Netto JM, Xue A, Tchkonia T, Kirkland JL, Seshadri S, Salardini A, Musi N, Bateman RJ, Gonzales MM, Orr ME. Evaluation of Exploratory Fluid Biomarker Results from a Phase 1 Senolytic Trial in Mild Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-3994894. [PMID: 38496619 PMCID: PMC10942554 DOI: 10.21203/rs.3.rs-3994894/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Senescent cell accumulation contributes to the progression of age-related disorders including Alzheimer's disease (AD). Clinical trials evaluating senolytics, drugs that clear senescent cells, are underway, but lack standardized outcome measures. Our team recently published data from the first open-label trial to evaluate senolytics (dasatinib plus quercetin) in AD. After 12-weeks of intermittent treatment, we reported brain exposure to dasatinib, favorable safety and tolerability, and modest post-treatment changes in cerebrospinal fluid (CSF) inflammatory and AD biomarkers using commercially available assays. Herein, we present more comprehensive exploratory analyses of senolytic associated changes in AD relevant proteins, metabolites, lipids, and transcripts measured across blood, CSF, and urine. These analyses included mass spectrometry for precise quantification of amyloid beta (Aß) and tau in CSF; immunoassays to assess senescence associated secretory factors in plasma, CSF, and urine; mass spectrometry analysis of urinary metabolites and lipids in blood and CSF; and transcriptomic analyses relevant to chronic stress measured in peripheral blood cells. Levels of Aß and tau species remained stable. Targeted cytokine and chemokine analyses revealed treatment-associated increases in inflammatory plasma fractalkine and MMP-7 and CSF IL-6. Urinary metabolites remained unchanged. Modest treatment-associated lipid profile changes suggestive of decreased inflammation were observed both peripherally and centrally. Blood transcriptomic analysis indicated downregulation of inflammatory genes including FOS, FOSB, IL1β, IL8, JUN, JUNB, PTGS2. These data provide a foundation for developing standardized outcome measures across senolytic studies and indicate distinct biofluid-specific signatures that will require validation in future studies. ClinicalTrials.gov: NCT04063124.
Collapse
Affiliation(s)
- Valentina R. Garbarino
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Juan Pablo Palavicini
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Justin Melendez
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Nicolas Barthelemy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Yingxin He
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Tiffany F. Kautz
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Marisa Lopez-Cruzan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Julia J. Mathews
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Peng Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Afaf Saliba
- Center for Precision Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Nagarjunachary Ragi
- Center for Precision Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kumar Sharma
- Center for Precision Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Suzanne Craft
- Department of Internal Medicine Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | - Ailing Xue
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Tamara Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | | | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Arash Salardini
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Neurology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Nicolas Musi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Randall J. Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Tracy Family SILQ Center for Neurodegenerative Biology, St. Louis, MO, USA
| | - Mitzi M. Gonzales
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Neurology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Miranda E. Orr
- Department of Internal Medicine Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Salisbury VA Medical Center, Salisbury, NC, 28144, USA
| |
Collapse
|
13
|
Mosconi L, Williams S, Carlton C, Zarate C, Boneu C, Fauci F, Ajila T, Nerattini M, Jett S, Andy C, Battista M, Pahlajani S, Osborne J, Brinton RD, Dyke JP. Sex-specific associations of serum cortisol with brain biomarkers of Alzheimer's risk. Sci Rep 2024; 14:5519. [PMID: 38448497 PMCID: PMC10918173 DOI: 10.1038/s41598-024-56071-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024] Open
Abstract
Emerging evidence implicates chronic psychological stress as a risk factor for Alzheimer's disease (AD). Herein, we examined the relationships between serum cortisol and multimodality brain AD biomarkers in 277 cognitively normal midlife individuals at risk for AD. Overall, higher cortisol was associated with lower total brain volume, lower glucose metabolism (CMRglc) in frontal cortex, and higher β-amyloid (Aβ) load in AD-vulnerable regions; and marginally associated with phosphocreatine to ATP ratios (PCr/ATP) in precuneus and parietal regions. Sex-specific modification effects were noted: in women, cortisol exhibited stronger associations with Aβ load and frontal CMRglc, the latter being more pronounced postmenopause. In men, cortisol exhibited stronger associations with gray matter volume and PCr/ATP measures. Higher cortisol was associated with poorer delayed memory in men but not in women. Results were adjusted for age, Apolipoprotein E (APOE) epsilon 4 status, midlife health factors, and hormone therapy use. These results suggest sex-specific neurophysiological responses to stress, and support a role for stress reduction in AD prevention.
Collapse
Affiliation(s)
- Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA.
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA.
| | - Schantel Williams
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Caroline Carlton
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Camila Zarate
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Camila Boneu
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Francesca Fauci
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Trisha Ajila
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Matilde Nerattini
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
- Department of Clinical Pathophysiology, Nuclear Medicine Unit, University of Florence, Florence, Italy
| | - Steven Jett
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Michael Battista
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Joseph Osborne
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Roberta Diaz Brinton
- Department of Neurology and Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Jonathan P Dyke
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
14
|
Armenta-Castro A, Núñez-Soto MT, Rodriguez-Aguillón KO, Aguayo-Acosta A, Oyervides-Muñoz MA, Snyder SA, Barceló D, Saththasivam J, Lawler J, Sosa-Hernández JE, Parra-Saldívar R. Urine biomarkers for Alzheimer's disease: A new opportunity for wastewater-based epidemiology? ENVIRONMENT INTERNATIONAL 2024; 184:108462. [PMID: 38335627 DOI: 10.1016/j.envint.2024.108462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 01/16/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024]
Abstract
While Alzheimer's disease (AD) diagnosis, management, and care have become priorities for healthcare providers and researcher's worldwide due to rapid population aging, epidemiologic surveillance efforts are currently limited by costly, invasive diagnostic procedures, particularly in low to middle income countries (LMIC). In recent years, wastewater-based epidemiology (WBE) has emerged as a promising tool for public health assessment through detection and quantification of specific biomarkers in wastewater, but applications for non-infectious diseases such as AD remain limited. This early review seeks to summarize AD-related biomarkers and urine and other peripheral biofluids and discuss their potential integration to WBE platforms to guide the first prospective efforts in the field. Promising results have been reported in clinical settings, indicating the potential of amyloid β, tau, neural thread protein, long non-coding RNAs, oxidative stress markers and other dysregulated metabolites for AD diagnosis, but questions regarding their concentration and stability in wastewater and the correlation between clinical levels and sewage circulation must be addressed in future studies before comprehensive WBE systems can be developed.
Collapse
Affiliation(s)
| | - Mónica T Núñez-Soto
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico
| | - Kassandra O Rodriguez-Aguillón
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico; Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| | - Alberto Aguayo-Acosta
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico; Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| | - Mariel Araceli Oyervides-Muñoz
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico; Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| | - Shane A Snyder
- Nanyang Environment & Water Research Institute (NEWRI), Nanyang Technological University, Singapore
| | - Damià Barceló
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research, IDAEA-CSIC, Jordi Girona, 18-26, 08034 Barcelona, Spain; Sustainability Cluster, School of Engineering at the UPES, Dehradun, Uttarakhand, India
| | - Jayaprakash Saththasivam
- Water Center, Qatar Environment & Energy Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Qatar
| | - Jenny Lawler
- Water Center, Qatar Environment & Energy Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Qatar
| | - Juan Eduardo Sosa-Hernández
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico; Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico.
| | - Roberto Parra-Saldívar
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey 64849, Mexico; Tecnologico de Monterrey, Institute of Advanced Materials for Sustainable Manufacturing, Monterrey 64849, Mexico
| |
Collapse
|
15
|
Gandy HM, Hollis F, Hernandez CM, McQuail JA. Aging or chronic stress impairs working memory and modulates GABA and glutamate gene expression in prelimbic cortex. Front Aging Neurosci 2024; 15:1306496. [PMID: 38259638 PMCID: PMC10800675 DOI: 10.3389/fnagi.2023.1306496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
The glucocorticoid (GC) hypothesis posits that effects of stress and dysregulated hypothalamic-pituitary-adrenal axis activity accumulate over the lifespan and contribute to impairment of neural function and cognition in advanced aging. The validity of the GC hypothesis is bolstered by a wealth of studies that investigate aging of the hippocampus and decline of associated mnemonic functions. The prefrontal cortex (PFC) mediates working memory which also decreases with age. While the PFC is susceptible to stress and GCs, few studies have formally assessed the application of the GC hypothesis to PFC aging and working memory. Using parallel behavioral and molecular approaches, we compared the effects of normal aging versus chronic variable stress (CVS) on working memory and expression of genes that encode for effectors of glutamate and GABA signaling in male F344 rats. Using an operant delayed match-to-sample test of PFC-dependent working memory, we determined that normal aging and CVS each significantly impaired mnemonic accuracy and reduced the total number of completed trials. We then determined that normal aging increased expression of Slc6a11, which encodes for GAT-3 GABA transporter expressed by astrocytes, in the prelimbic (PrL) subregion of the PFC. CVS increased PrL expression of genes associated with glutamatergic synapses: Grin2b that encodes the GluN2B subunit of NMDA receptor, Grm4 that encodes for metabotropic glutamate receptor 4 (mGluR4), and Plcb1 that encodes for phospholipase C beta 1, an intracellular signaling enzyme that transduces signaling of Group I mGluRs. Beyond the identification of specific genes that were differentially expressed between the PrL in normal aging or CVS, examination of Log2 fold-changes for all expressed glutamate and GABA genes revealed a positive association between molecular phenotypes of aging and CVS in the PrL but no association in the infralimbic subregion. Consistent with predictions of the GC hypothesis, PFC-dependent working memory and PrL glutamate/GABA gene expression demonstrate comparable sensitivity to aging and chronic stress. However, changes in expression of specific genes affiliated with regulation of extracellular GABA in normal aging vs. genes encoding for effectors of glutamatergic signaling during CVS suggest the presence of unique manifestations of imbalanced inhibitory and excitatory signaling in the PFC.
Collapse
Affiliation(s)
- Hannah M. Gandy
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Fiona Hollis
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Columbia VA Health Care System, Columbia, SC, United States
| | - Caesar M. Hernandez
- Department of Medicine, Division of Gerontology, Geriatrics, and Palliative Care, The University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Joseph A. McQuail
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| |
Collapse
|
16
|
Steinbach MJ, Denburg NL. Melatonin in Alzheimer's Disease: Literature Review and Therapeutic Trials. J Alzheimers Dis 2024; 101:S193-S204. [PMID: 39422936 DOI: 10.3233/jad-230760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
There are currently no effective treatments to prevent, halt, or reverse Alzheimer's disease (AD), the most common cause of dementia in older adults. Melatonin, a relatively harmless over-the-counter supplement, may offer some benefits to patients with AD. Melatonin is known for its sleep-enhancing properties, but research shows that it may provide other advantages as well, such as antioxidant and anti-amyloidogenic properties. Clinical trials for melatonin use in AD have mixed results but, overall, show modest benefits. However, it is difficult to interpret clinical research in this area as there is little standardization to guide the administration and study of melatonin. This review covers basic biology and clinical research on melatonin in AD focusing on prominent hypotheses of pathophysiology of neurodegeneration and cognitive decline in AD (i.e., amyloid and tau hypotheses, antioxidant and anti-inflammation, insulin resistance and glucose homeostasis, the cholinergic hypothesis, sleep regulation, and the hypothalamic-pituitary-adrenal axis and cortisol). This is followed by a discussion on pending clinical trials, considerations for future research protocols, and open questions in the field.
Collapse
Affiliation(s)
- Marilyn J Steinbach
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- Department of Neurology, Division of Cognitive Neuroscience, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Natalie L Denburg
- Department of Neurology, Division of Cognitive Neuroscience, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
17
|
Ahmed SB, Ahmad S, Pan H. Case Report and Literature Review of an Atypical Polymyalgia Rheumatica and Its Management. Int Med Case Rep J 2023; 16:873-885. [PMID: 38163043 PMCID: PMC10757773 DOI: 10.2147/imcrj.s440486] [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: 09/15/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024] Open
Abstract
Polymyalgia rheumatica (PMR) is a systemic inflammatory disease of the elderly population that increases in incidence as age advances. It is characterised by the sudden or sub-acute onset of symptoms affecting the shoulder and pelvic girdles, often accompanied by constitutional symptoms. Due to the lack of consensual diagnostic criteria and specific laboratory or radiological investigations for PMR, its diagnosis can be very challenging, particularly because it can be mimicked or masked by other geriatric syndromes. PMR responds well to glucocorticoid treatment, but if left untreated, can lead to morbidity and poor quality of life. We present the case of an 87-year-old male who presented with a one-week history of localised pain in the left hip joint, later involving the contralateral hip. Previously able to ambulate unaided, his mobility was now severely impaired. Due to his Alzheimer's dementia and multiple comorbid geriatric conditions, extensive investigations were undertaken before a diagnosis of atypical PMR was reached. Treatment with a low dose of prednisolone led to a full recovery. This case highlights the inconsistency between an atypical presentation and the classic presentation of PMR and draws attention to the possibility of missed diagnosis in older, frail patients. Atypical symptomatology on top of cognitive impairment and language barriers can be easily overlooked and left untreated and could lead to severe adverse outcomes. Accurate diagnosis is crucial, as PMR is readily diagnosed, but the treatment with glucocorticoids, though generally straightforward, can pose challenges, particularly when dealing with polypharmacy and multiple coexisting health conditions.
Collapse
Affiliation(s)
- Saad Bilal Ahmed
- Monash Health Rehabilitation and Aged Care Services, Melbourne, Australia
| | - Saara Ahmad
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Hanmei Pan
- Monash Health Rehabilitation and Aged Care Services, Melbourne, Australia
| |
Collapse
|
18
|
Aizpurua-Perez I, Arregi A, Labaka A, Martinez-Villar A, Perez-Tejada J. Psychological resilience and cortisol levels in adults: A systematic review. Am J Hum Biol 2023; 35:e23954. [PMID: 37395446 DOI: 10.1002/ajhb.23954] [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: 11/17/2022] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/04/2023] Open
Abstract
Resilience or the capacity to "bend but not break" refers to the ability to maintain or regain psychobiological equilibrium during or after exposure to stressful life events. Specifically, resilience has been proposed as a potential resource for staving off pathological states that often emerge after exposure to repeated stress and that are related to alterations in circulating cortisol. The aim of this systematic review of the literature was to gather evidence related to the relationship between psychological resilience and cortisol levels in adult humans. An extensive systematic search was carried out following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) method in the PubMed and Web of Science databases. In total, 1256 articles were identified and, of these, 35 peer-reviewed articles were included in the systematic review. We categorized findings according to (1) the short and long-term secretion period covered by the cortisol matrices selected by studies and also according to (2) the differentiated diurnal, phasic (acute), and tonic (basal) components of the HPA output to which they refer and their relationships with resilience. Reported relationships between psychological resilience and distinct cortisol output parameters varied widely across studies, finding positive, negative, and null associations between the two variables. Notably, several of the studies that found no relationship between resilience and cortisol used a single morning saliva or plasma sample as their assessment of HPA axis activity. Despite limitations such as the great variability of the instruments and methods used by the studies to measure both resilience and cortisol, together with their high heterogeneity and small sample sizes, the evidence found in this systematic review points to the potential of resilience as a modifiable key factor to modulate the physiological response to stress. Therefore, further exploration of the interaction between the two variables is necessary for the eventual development of future interventions aimed at promoting resilience as an essential component of health prevention.
Collapse
Affiliation(s)
- Ibane Aizpurua-Perez
- Department of Basic Psychological Processes and their Development, University of the Basque Country, San Sebastian, Spain
| | - Amaia Arregi
- Department of Basic Psychological Processes and their Development, University of the Basque Country, San Sebastian, Spain
| | - Ainitze Labaka
- Department of Nursing II, University of the Basque Country, San Sebastian, Spain
| | | | | |
Collapse
|
19
|
Warde KM, Smith LJ, Basham KJ. Age-related Changes in the Adrenal Cortex: Insights and Implications. J Endocr Soc 2023; 7:bvad097. [PMID: 37564884 PMCID: PMC10410302 DOI: 10.1210/jendso/bvad097] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Indexed: 08/12/2023] Open
Abstract
Aging is characterized by a gradual decline in physiological function. This process affects all organs including the adrenal cortex, which normally functions to produce essential steroid hormones including mineralocorticoids, glucocorticoids, and androgens. With increasing age, features such as reduced adrenal cortex size, altered zonation, and increased myeloid immune cell infiltration substantially alter the structure and function of the adrenal cortex. Many of these hallmark features of adrenal cortex aging occur both in males and females, yet are more enhanced in males. Hormonally, a substantial reduction in adrenal androgens is a key feature of aging, which is accompanied by modest changes in aldosterone and cortisol. These hormonal changes are associated with various pathological consequences including impaired immune responses, decreased bone health, and accelerated age-related diseases. One of the most notable changes with adrenal aging is the increased incidence of adrenal tumors, which is sex dimorphic with a higher prevalence in females. Increased adrenal tumorigenesis with age is likely driven by both an increase in genetic mutations as well as remodeling of the tissue microenvironment. Novel antiaging strategies offer a promising avenue to mitigate adrenal aging and alleviate age-associated pathologies, including adrenal tumors.
Collapse
Affiliation(s)
- Kate M Warde
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Lorenzo J Smith
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
20
|
Luo J, Beam CR, Gatz M. Is Stress an Overlooked Risk Factor for Dementia? A Systematic Review from a Lifespan Developmental Perspective. PREVENTION SCIENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR PREVENTION RESEARCH 2023; 24:936-949. [PMID: 35622193 DOI: 10.1007/s11121-022-01385-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/26/2022]
Abstract
Stress exposure and stress reactivity may be potent factors associated with increased risk of dementia. The 2017 Lancet Commission on Dementia and its 2020 update reviewed modifiable risk factors associated with dementia, but stress was not addressed directly. The present study provides a focused review of the association between stress and dementia across the lifespan, with measures of stress including stress exposure, psychological stress, posttraumatic stress disorder (PTSD), and biological markers of stress. Published research articles were identified in the American Psychological Association PsycINFO database (1887-2021), Web of Science database, and Google Scholar. A total of 53 samples from 40 studies published from 1985 to 2020 met inclusion criteria. Results suggest that stressful life events that occur earlier in the lifespan, such as loss of a parent, psychological stress experienced in midlife, and extreme stress responses, i.e., PTSD, correlate with higher risk of dementia. Although results generally are mixed, a consistent theme is that stress experienced earlier in the lifespan and chronic stress portend the greatest risk of dementia. Reducing stress exposure and improving stress management when stress exposure cannot be changed are thus relevant strategies in dementia risk reduction.
Collapse
Affiliation(s)
- Jing Luo
- Department of Medical Social Sciences, Feinberg School of Medicine, Northwestern University, 625 N. Michigan Avenue, Chicago, IL, 60611, USA.
| | - Christopher R Beam
- Department of Psychology, University of Southern California, Los Angeles, CA, 90089-1061, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Margaret Gatz
- Department of Psychology, University of Southern California, Los Angeles, CA, 90089-1061, USA
- Center for Economic and Social Research, University of Southern California, Los Angeles, CA, 90089-3332, USA
| |
Collapse
|
21
|
Wu C, Feng Y. Exploring the potential of mindfulness-based therapy in the prevention and treatment of neurodegenerative diseases based on molecular mechanism studies. Front Neurosci 2023; 17:1097067. [PMID: 37383106 PMCID: PMC10293639 DOI: 10.3389/fnins.2023.1097067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 05/03/2023] [Indexed: 06/30/2023] Open
Abstract
Neurodegenerative diseases (ND) have received increasing attention due to their irreversibility, but there is still no means to completely cure ND in clinical practice. Mindfulness therapy (MT), including Qigong, Tai Chi, meditation, and yoga, etc., has become an effective complementary treatment modality in solving clinical and subclinical problems due to its advantages of low side effects, less pain, and easy acceptance by patients. MT is primarily used to treat mental and emotional disorders. In recent years, evidence has shown that MT has a certain therapeutic effect on ND with a potential molecular basis. In this review, we summarize the pathogenesis and risk factors of Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), relating to telomerase activity, epigenetics, stress, and the pro-inflammatory transcription factor nuclear factor kappa B (NF-κB) mediated inflammatory response, and analyze the molecular mechanism basis of MT to prevent and treat ND, to provide possible explanations for the potential of MT treatments for ND.
Collapse
|
22
|
Biagetti B, Puig-Domingo M. Age-Related Hormones Changes and Its Impact on Health Status and Lifespan. Aging Dis 2023; 14:605-620. [PMID: 37191429 PMCID: PMC10187696 DOI: 10.14336/ad.2022.1109] [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: 10/11/2022] [Accepted: 11/09/2022] [Indexed: 05/17/2023] Open
Abstract
The increase in life expectancy is accompanied with an increased consultation of age-related pathologies including endocrine disorders. Two main areas are focusing the attention of medical and social research in older population: the diagnosis and care of this heterogeneous population, and the interventional measures potentially useful to mitigate age-related functional declines and to increase health and quality of lifespan. Thus, better understanding the physiopathology of aging and establishing accurate diagnostic and personalized approaches are a priority and currently an unmet need of the medical community. The endocrine system plays a major role in survival and lifespan through regulating vital processes such as energy consumption and optimizing the stress response among others. The aim of this paper is to review the physiological evolution of the main hormonal functions in aging and its clinical translation to improve our approach to the aging patient.
Collapse
Affiliation(s)
- Betina Biagetti
- Endocrinology & Nutrition Service, Vall d’Hebron University Hospital and Vall d'Hebron Research Institute (VHIR), Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain.
| | - Manel Puig-Domingo
- Endocrinology & Nutrition Service, Germans Trias Hospital and Research Institute, Badalona, Department of Medicine, Autonomous University of Barcelona, Badalona, Spain.
| |
Collapse
|
23
|
Dronse J, Ohndorf A, Richter N, Bischof GN, Fassbender R, Behfar Q, Gramespacher H, Dillen K, Jacobs HIL, Kukolja J, Fink GR, Onur OA. Serum cortisol is negatively related to hippocampal volume, brain structure, and memory performance in healthy aging and Alzheimer's disease. Front Aging Neurosci 2023; 15:1154112. [PMID: 37251803 PMCID: PMC10213232 DOI: 10.3389/fnagi.2023.1154112] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/19/2023] [Indexed: 05/31/2023] Open
Abstract
Objective Elevated cortisol levels have been frequently reported in Alzheimer's disease (AD) and linked to brain atrophy, especially of the hippocampus. Besides, high cortisol levels have been shown to impair memory performance and increase the risk of developing AD in healthy individuals. We investigated the associations between serum cortisol levels, hippocampal volume, gray matter volume and memory performance in healthy aging and AD. Methods In our cross-sectional study, we analyzed the relationships between morning serum cortisol levels, verbal memory performance, hippocampal volume, and whole-brain voxel-wise gray matter volume in an independent sample of 29 healthy seniors (HS) and 29 patients along the spectrum of biomarker-based AD. Results Cortisol levels were significantly elevated in patients with AD as compared to HS, and higher cortisol levels were correlated with worse memory performance in AD. Furthermore, higher cortisol levels were significantly associated with smaller left hippocampal volumes in HS and indirectly negatively correlated to memory function through hippocampal volume. Higher cortisol levels were further related to lower gray matter volume in the hippocampus and temporal and parietal areas in the left hemisphere in both groups. The strength of this association was similar in HS and AD. Conclusion In AD, cortisol levels are elevated and associated with worse memory performance. Furthermore, in healthy seniors, higher cortisol levels show a detrimental relationship with brain regions typically affected by AD. Thus, increased cortisol levels seem to be indirectly linked to worse memory function even in otherwise healthy individuals. Cortisol may therefore not only serve as a biomarker of increased risk for AD, but maybe even more importantly, as an early target for preventive and therapeutic interventions.
Collapse
Affiliation(s)
- Julian Dronse
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anna Ohndorf
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nils Richter
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gérard N. Bischof
- Department of Nuclear Medicine, Multimodal Neuroimaging Group, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ronja Fassbender
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Qumars Behfar
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hannes Gramespacher
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Kim Dillen
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Palliative Medicine, Multimodal Neuroimaging Group, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Heidi I. L. Jacobs
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, Netherlands
| | - Juraj Kukolja
- Department of Neurology and Clinical Neurophysiology, Helios University Hospital Wuppertal, Wuppertal, Germany
- Faculty of Health Witten/Herdecke University, Witten, Germany
| | - Gereon R. Fink
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Oezguer A. Onur
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
24
|
Ren Y, Savadlou A, Park S, Siska P, Epp JR, Sargin D. The impact of loneliness and social isolation on the development of cognitive decline and Alzheimer's Disease. Front Neuroendocrinol 2023; 69:101061. [PMID: 36758770 DOI: 10.1016/j.yfrne.2023.101061] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/19/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Alzheimer's Disease (AD) is the leading cause of dementia, observed at a higher incidence in women compared with men. Treatments aimed at improving pathology in AD remain ineffective to stop disease progression. This makes the detection of the early intervention strategies to reduce future disease risk extremely important. Isolation and loneliness have been identified among the major risk factors for AD. The increasing prevalence of both loneliness and AD emphasizes the urgent need to understand this association to inform treatment. Here we present a comprehensive review of both clinical and preclinical studies that investigated loneliness and social isolation as risk factors for AD. We discuss that understanding the mechanisms of how loneliness exacerbates cognitive impairment and AD with a focus on sex differences will shed the light for the underlying mechanisms regarding loneliness as a risk factor for AD and to develop effective prevention or treatment strategies.
Collapse
Affiliation(s)
- Yi Ren
- Department of Cell Biology and Anatomy, University of Calgary, Canada; Cumming School of Medicine, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada
| | - Aisouda Savadlou
- Department of Psychology, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Canada
| | - Soobin Park
- Department of Psychology, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Canada
| | - Paul Siska
- Department of Psychology, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Canada
| | - Jonathan R Epp
- Department of Cell Biology and Anatomy, University of Calgary, Canada; Cumming School of Medicine, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada
| | - Derya Sargin
- Department of Psychology, University of Calgary, Canada; Department of Physiology & Pharmacology, University of Calgary, Canada; Hotchkiss Brain Institute, University of Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Canada.
| |
Collapse
|
25
|
Rao RV, Subramaniam KG, Gregory J, Bredesen AL, Coward C, Okada S, Kelly L, Bredesen DE. Rationale for a Multi-Factorial Approach for the Reversal of Cognitive Decline in Alzheimer's Disease and MCI: A Review. Int J Mol Sci 2023; 24:ijms24021659. [PMID: 36675177 PMCID: PMC9865291 DOI: 10.3390/ijms24021659] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial, progressive, neurodegenerative disease typically characterized by memory loss, personality changes, and a decline in overall cognitive function. Usually manifesting in individuals over the age of 60, this is the most prevalent type of dementia and remains the fifth leading cause of death among Americans aged 65 and older. While the development of effective treatment and prevention for AD is a major healthcare goal, unfortunately, therapeutic approaches to date have yet to find a treatment plan that produces long-term cognitive improvement. Drugs that may be able to slow down the progression rate of AD are being introduced to the market; however, there has been no previous solution for preventing or reversing the disease-associated cognitive decline. Recent studies have identified several factors that contribute to the progression and severity of the disease: diet, lifestyle, stress, sleep, nutrient deficiencies, mental health, socialization, and toxins. Thus, increasing evidence supports dietary and other lifestyle changes as potentially effective ways to prevent, slow, or reverse AD progression. Studies also have demonstrated that a personalized, multi-therapeutic approach is needed to improve metabolic abnormalities and AD-associated cognitive decline. These studies suggest the effects of abnormalities, such as insulin resistance, chronic inflammation, hypovitaminosis D, hormonal deficiencies, and hyperhomocysteinemia, in the AD process. Therefore a personalized, multi-therapeutic program based on an individual's genetics and biochemistry may be preferable over a single-drug/mono-therapeutic approach. This article reviews these multi-therapeutic strategies that identify and attenuate all the risk factors specific to each affected individual. This article systematically reviews studies that have incorporated multiple strategies that target numerous factors simultaneously to reverse or treat cognitive decline. We included high-quality clinical trials and observational studies that focused on the cognitive effects of programs comprising lifestyle, physical, and mental activity, as well as nutritional aspects. Articles from PubMed Central, Scopus, and Google Scholar databases were collected, and abstracts were reviewed for relevance to the subject matter. Epidemiological, pathological, toxicological, genetic, and biochemical studies have all concluded that AD represents a complex network insufficiency. The research studies explored in this manuscript confirm the need for a multifactorial approach to target the various risk factors of AD. A single-drug approach may delay the progression of memory loss but, to date, has not prevented or reversed it. Diet, physical activity, sleep, stress, and environment all contribute to the progression of the disease, and, therefore, a multi-factorial optimization of network support and function offers a rational therapeutic strategy. Thus, a multi-therapeutic program that simultaneously targets multiple factors underlying the AD network may be more effective than a mono-therapeutic approach.
Collapse
Affiliation(s)
- Rammohan V. Rao
- Apollo Health, Burlingame, CA 94011, USA
- Correspondence: (R.V.R.); (D.E.B.)
| | | | | | | | | | - Sho Okada
- Apollo Health, Burlingame, CA 94011, USA
| | | | - Dale E. Bredesen
- Apollo Health, Burlingame, CA 94011, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90024, USA
- Correspondence: (R.V.R.); (D.E.B.)
| |
Collapse
|
26
|
Campos HC, Ribeiro DE, Hashiguchi D, Glaser T, Milanis MDS, Gimenes C, Suchecki D, Arida RM, Ulrich H, Monteiro Longo B. Neuroprotective effects of resistance physical exercise on the APP/PS1 mouse model of Alzheimer's disease. Front Neurosci 2023; 17:1132825. [PMID: 37090809 PMCID: PMC10116002 DOI: 10.3389/fnins.2023.1132825] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/16/2023] [Indexed: 04/25/2023] Open
Abstract
Introduction Physical exercise has beneficial effects by providing neuroprotective and anti-inflammatory responses to AD. Most studies, however, have been conducted with aerobic exercises, and few have investigated the effects of other modalities that also show positive effects on AD, such as resistance exercise (RE). In addition to its benefits in developing muscle strength, balance and muscular endurance favoring improvements in the quality of life of the elderly, RE reduces amyloid load and local inflammation, promotes memory and cognitive improvements, and protects the cortex and hippocampus from the degeneration that occurs in AD. Similar to AD patients, double-transgenic APPswe/PS1dE9 (APP/PS1) mice exhibit Αβ plaques in the cortex and hippocampus, hyperlocomotion, memory deficits, and exacerbated inflammatory response. Therefore, the aim of this study was to investigate the effects of 4 weeks of RE intermittent training on the prevention and recovery from these AD-related neuropathological conditions in APP/PS1 mice. Methods For this purpose, 6-7-month-old male APP/PS1 transgenic mice and their littermates, negative for the mutations (CTRL), were distributed into three groups: CTRL, APP/PS1, APP/PS1+RE. RE training lasted four weeks and, at the end of the program, the animals were tested in the open field test for locomotor activity and in the object recognition test for recognition memory evaluation. The brains were collected for immunohistochemical analysis of Aβ plaques and microglia, and blood was collected for plasma corticosterone by ELISA assay. Results APP/PS1 transgenic sedentary mice showed increased hippocampal Aβ plaques and higher plasma corticosterone levels, as well as hyperlocomotion and reduced central crossings in the open field test, compared to APP/PS1 exercised and control animals. The intermittent program of RE was able to recover the behavioral, corticosterone and Aβ alterations to the CTRL levels. In addition, the RE protocol increased the number of microglial cells in the hippocampus of APP/PS1 mice. Despite these alterations, no memory impairment was observed in APP/PS1 mice in the novel object recognition test. Discussion Altogether, the present results suggest that RE plays a role in alleviating AD symptoms, and highlight the beneficial effects of RE training as a complementary treatment for AD.
Collapse
Affiliation(s)
- Henrique Correia Campos
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deidiane Elisa Ribeiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Debora Hashiguchi
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- Instituto do Cérebro - ICe, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Milena da Silva Milanis
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Christiane Gimenes
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Deborah Suchecki
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ricardo Mario Arida
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- *Correspondence: Henning Ulrich, ; Beatriz Monteiro Longo, ;
| | - Beatriz Monteiro Longo
- Laboratory of Neurophysiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
- *Correspondence: Henning Ulrich, ; Beatriz Monteiro Longo, ;
| |
Collapse
|
27
|
Minné D, Marnewick JL, Engel-Hills P. Early Chronic Stress Induced Changes within the Locus Coeruleus in Sporadic Alzheimer's Disease. Curr Alzheimer Res 2023; 20:301-317. [PMID: 37872793 DOI: 10.2174/1567205020666230811092956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 10/25/2023]
Abstract
Chronic exposure to stress throughout the lifespan has been the focus of many studies on Alzheimer's disease (AD) because of the similarities between the biological mechanisms involved in chronic stress and the pathophysiology of AD. In fact, the earliest abnormality associated with the disease is the presence of phosphorylated tau protein in locus coeruleus neurons, a brain structure highly responsive to stress and perceived threat. Here, we introduce allostatic load as a useful concept for understanding many of the complex, interacting neuropathological changes involved in the AD degenerative process. In response to chronic stress, aberrant tau proteins that begin to accumulate within the locus coeruleus decades prior to symptom onset appear to represent a primary pathological event in the AD cascade, triggering a wide range of interacting brain changes involving neuronal excitotoxicity, endocrine alterations, inflammation, oxidative stress, and amyloid plaque exacerbation. While it is acknowledged that stress will not necessarily be the major precipitating factor in all cases, early tau-induced changes within the locus coeruleus-norepinephrine pathway suggests that a therapeutic window might exist for preventative measures aimed at managing stress and restoring balance within the HPA axis.
Collapse
Affiliation(s)
- Donné Minné
- Applied Microbial & Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
- Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| | - Jeanine L Marnewick
- Applied Microbial & Health Biotechnology Institute, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| | - Penelope Engel-Hills
- Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Cape Town, 7535, South Africa
| |
Collapse
|
28
|
Lv YN, Cui Y, Zhang B, Huang SM. Sleep deficiency promotes Alzheimer's disease development and progression. Front Neurol 2022; 13:1053942. [PMID: 36588906 PMCID: PMC9795181 DOI: 10.3389/fneur.2022.1053942] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Sleep disorders are a common health problem in modern society. Long-term sleep deficiency increases the risk for Alzheimer's disease. However, the exact mechanisms by which sleep deficiency affects Alzheimer's disease remain unclear. Therefore, we reviewed the relevant studies and investigated the role of sleep deprivation in Alzheimer's disease pathogenesis. Sleep deficiency was found to be associated with oxidative stress, β-amyloid protein deposition, tau hyperphosphorylation, and neuroinflammation, which are known to increase the risk for Alzheimer's disease. In addition, insufficient sleep also increases glucocorticoid levels, decreases brain-derived neurotrophic factor levels, and reduces the number of synapses in the central nervous system. These factors also promote Alzheimer's disease development and progression. The present study showed that a growing body of evidence supports an association between sleep disturbances and Alzheimer's disease. It discusses the role of sleep insufficiency in Alzheimer's disease pathogenesis, which may provide a theoretical basis for effective treatment and prevention strategies.
Collapse
Affiliation(s)
- Ya-Nan Lv
- Department of Neuroscience, Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Cui
- Department of Veterinary Medicine, School of Animal Science and Technology, Hainan University, Haikou, China
| | - Bo Zhang
- Department of Neuroscience, Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China,*Correspondence: Bo Zhang
| | - Shu-Ming Huang
- Department of Neuroscience, Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
29
|
Development of Post-Stroke Cognitive and Depressive Disturbances: Associations with Neurohumoral Indices. Curr Issues Mol Biol 2022; 44:6290-6305. [PMID: 36547090 PMCID: PMC9776624 DOI: 10.3390/cimb44120429] [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: 11/06/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Neuropsychiatric complications, in particular cognitive and depressive disorders, are common consequences of ischemic stroke (IS) and complicate the rehabilitation, quality of life, and social adaptation of patients. The hypothalamic-pituitary-adrenal (HPA) system, sympathoadrenal medullary system (SAMS), and inflammatory processes are believed to be involved in the pathogenesis of these disorders. This study aimed to explore these systems in IS patients, including those with post-stroke cognitive and depressive disorders, within a year after IS. Indices of the HPA axis, inflammatory system, and SAMS were measured in blood serum (cortisol, interleukin-6 (IL-6)), plasma (adrenocorticotropic hormone), and saliva (cortisol, α-amylase). During one year after mild/moderate IS (NIHSS score 5.9 ± 4.3), serum cortisol and salivary α-amylase levels remained elevated in the total cohort. In the group with further cognitive decline, serum and salivary cortisol levels were elevated during the acute period of IS. In the group with poststroke depressive disorder, salivary α-amylase was constantly elevated, while serum IL-6 was minimal during the acute period. The results suggest prolonged hyperactivation of the HPA axis and SAMS after IS. Specifically, post-stroke cognitive impairment was associated with hyperactivation of the HPA axis during the acute IS period, while post-stroke depressive disorder was associated with the chronic inflammatory process and hyperactivation of SAMS during the follow-up period.
Collapse
|
30
|
Bayer TA, Van Patten R, Hershkowitz D, Epstein-Lubow G, Rudolph JL. Comorbidity and Management of Concurrent Psychiatric and Medical Disorders. Psychiatr Clin North Am 2022; 45:745-763. [PMID: 36396277 DOI: 10.1016/j.psc.2022.07.006] [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] [Indexed: 11/15/2022]
Abstract
Aging increases susceptibility to medical and psychiatric comorbidity via interrelated biological, psychological, and social mechanisms. Mental status changes or other psychiatric symptoms occurring in older adults with medical disorders most often result from delirium, depression, or the onset of Alzheimer's disease and related dementias (ADRD). Clinicians can use evidence-based tools to evaluate such symptoms including the 4A's Test for delirium, the Saint Louis University Mental Status Exam, and the Geriatric Depression Scale. Innovative models such as collaborative care can improve the outcome of care of older adults with medical disorders requiring treatment for depression or ADRD..
Collapse
Affiliation(s)
- Thomas A Bayer
- Long-term Services and Supports Center of Innovation, Providence VA Medical Center, 353-373 Niagara St., Providence, RI 02907, USA; Division of Geriatrics and Palliative Medicine, Alpert Medical School of Brown University, 593 Eddy St., POB 438, Providence, RI 02903, USA.
| | - Ryan Van Patten
- Providence VA Medical Center, 830 Chalkstone Ave, Providence, RI 02908, USA; Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, 593 Eddy Street, APC9 Providence, RI 02903, USA
| | - Dylan Hershkowitz
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, 593 Eddy Street, APC9 Providence, RI 02903, USA
| | - Gary Epstein-Lubow
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, 593 Eddy Street, APC9 Providence, RI 02903, USA; Department of Health Services, Policy and Practice, Brown University School of Public Health, 121 S. Main Street, Providence, RI 02903, USA; Butler Hospital, 345 Blackstone Blvd, Providence, RI 02906, USA
| | - James L Rudolph
- Long-term Services and Supports Center of Innovation, Providence VA Medical Center, 353-373 Niagara St., Providence, RI 02907, USA; Division of Geriatrics and Palliative Medicine, Alpert Medical School of Brown University, 593 Eddy St., POB 438, Providence, RI 02903, USA; Department of Health Services, Policy and Practice, Brown University School of Public Health, 121 S. Main Street, Providence, RI 02903, USA
| |
Collapse
|
31
|
Iacobelli P. Circadian dysregulation and Alzheimer’s disease: A comprehensive review. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Alzheimer’s disease (AD), the foremost variant of dementia, has been associated with a menagerie of risk factors, many of which are considered to be modifiable. Among these modifiable risk factors is circadian rhythm, the chronobiological system that regulates sleep‐wake cycles, food consumption timing, hydration timing, and immune responses amongst many other necessary physiological processes. Circadian rhythm at the level of the suprachiasmatic nucleus (SCN), is tightly regulated in the human body by a host of biomolecular substances, principally the hormones melatonin, cortisol, and serotonin. In addition, photic information projected along afferent pathways to the SCN and peripheral oscillators regulates the synthesis of these hormones and mediates the manner in which they act on the SCN and its substructures. Dysregulation of this cycle, whether induced by environmental changes involving irregular exposure to light, or through endogenous pathology, will have a negative impact on immune system optimization and will heighten the deposition of Aβ and the hyperphosphorylation of the tau protein. Given these correlations, it appears that there is a physiologic association between circadian rhythm dysregulation and AD. This review will explore the physiology of circadian dysregulation in the AD brain, and will propose a basic model for its role in AD‐typical pathology, derived from the literature compiled and referenced throughout.
Collapse
Affiliation(s)
- Peter Iacobelli
- Department of Arts and Sciences, University of South Carolina, Columbia, USA
| |
Collapse
|
32
|
Aerqin Q, Wang ZT, Wu KM, He XY, Dong Q, Yu JT. Omics-based biomarkers discovery for Alzheimer's disease. Cell Mol Life Sci 2022; 79:585. [PMID: 36348101 DOI: 10.1007/s00018-022-04614-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorders presenting with the pathological hallmarks of amyloid plaques and tau tangles. Over the past few years, great efforts have been made to explore reliable biomarkers of AD. High-throughput omics are a technology driven by multiple levels of unbiased data to detect the complex etiology of AD, and it provides us with new opportunities to better understand the pathophysiology of AD and thereby identify potential biomarkers. Through revealing the interaction networks between different molecular levels, the ultimate goal of multi-omics is to improve the diagnosis and treatment of AD. In this review, based on the current AD pathology and the current status of AD diagnostic biomarkers, we summarize how genomics, transcriptomics, proteomics and metabolomics are all conducing to the discovery of reliable AD biomarkers that could be developed and used in clinical AD management.
Collapse
Affiliation(s)
- Qiaolifan Aerqin
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Kai-Min Wu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Xiao-Yu He
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
33
|
Shorey CL, Mulla RT, Mielke JG. The effects of synthetic glucocorticoid treatment for inflammatory disease on brain structure, function, and dementia outcomes: A systematic review. Brain Res 2022; 1798:148157. [DOI: 10.1016/j.brainres.2022.148157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/31/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
|
34
|
Sutin AR, Moffat SD, Resnick SM, Ferrucci L, Aschwanden D, Sesker AA, Luchetti M, Terracciano A. Five-Factor Model Personality Traits and 24-hour Urinary Cortisol in the Baltimore Longitudinal Study of Aging. Stress Health 2022; 38:837-843. [PMID: 35099848 PMCID: PMC9339027 DOI: 10.1002/smi.3130] [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: 08/11/2021] [Revised: 11/18/2021] [Accepted: 01/24/2022] [Indexed: 11/08/2022]
Abstract
Stress is implicated in models of personality and health as a mechanism that explains why traits like conscientiousness and neuroticism are associated with long-term health outcomes. Evidence for an association between personality and cortisol, a biological marker of stress, however, has been inconsistent. This study examined the association between Five-Factor Model personality traits and 24-h urinary cortisol (operationalised as a ratio of urinary free cortisol to creatinine) measured up to 12 times over intervals as long as 30 years in the Baltimore Longitudinal Study of Aging (Mage = 61.21, SD = 15.46; 49% female). There was a modest association between conscientiousness and lower mean-level cortisol that was attenuated only slightly in the fully-adjusted model. Neuroticism and the other traits were unrelated to cortisol levels, and none of the traits was related to cortisol change over time. The null association for neuroticism suggests that its relation with long-term health may be primarily through pathways other than cortisol. The modest association between conscientiousness and 24-h urinary cortisol replicates a previous finding with a longer-term measure of cortisol measured from hair, which calls for more research on the robustness and replicability of this finding. Cortisol may be one pathway through which conscientiousness is associated with health outcomes.
Collapse
Affiliation(s)
| | | | | | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health
| | | | | | | | | |
Collapse
|
35
|
Alonzo R, Anderson KK, Rodrigues R, Klar N, Chiodini P, Montero-Odasso M, Stranges S. Does Shiftwork Impact Cognitive Performance? Findings from the Canadian Longitudinal Study on Aging (CLSA). INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10124. [PMID: 36011754 PMCID: PMC9408351 DOI: 10.3390/ijerph191610124] [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: 07/14/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
Few large nationwide studies have investigated the relationship between shiftwork and cognitive performance, and little is known about whether and how psychological distress may impact this relationship. This study aimed to examine: (1) the cross-sectional relationship between shiftwork (yes/no) and some aspects of cognitive performance (declarative memory and executive functioning) and (2) the potential moderating effect of psychological distress among 20,610 community-dwelling adults from the comprehensive cohort of the Canadian Longitudinal Study on Aging (CLSA). Differences by sex and retirement status were also explored. Shiftwork was significantly associated with poorer performance for executive functioning (interference condition: ß = 0.47, 95% CI: 0.31 to 0.63; MAT: ß = -0.85, 95% CI: -1.21 to -0.50) but not for declarative memory. Completely and not/partly retired males showed poorer cognitive performance on executive functioning. However, no evidence of a moderating effect by psychological distress was found. Our findings confirm the association between shiftwork and cognitive performance and highlight important health correlates of shiftwork.
Collapse
Affiliation(s)
- Rea Alonzo
- Department of Epidemiology & Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Kelly K. Anderson
- Department of Epidemiology & Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
- Lawson Health Research Institute, London, ON N6C 2R5, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Rebecca Rodrigues
- Department of Epidemiology & Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Neil Klar
- Department of Epidemiology & Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Paolo Chiodini
- Medical Statistics Unit, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Manuel Montero-Odasso
- Department of Epidemiology & Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
- Gait and Brain Laboratory, Parkwood Institute, Lawson Health Research Institute, London, ON N6C 0A7, Canada
- Department of Medicine, Division of Geriatric Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Saverio Stranges
- Department of Epidemiology & Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
- Departments of Family Medicine and Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada
- Department of Precision Health, Luxembourg Institute of Health, Strassen, L-1445 Luxembourg, Luxembourg
| |
Collapse
|
36
|
Jimenez AG, Calderaro L, Clark S, Elacqua D, Hazen E, Lam V, Leightheiser GS. Can dogs serve as stress mediators to decrease salivary cortisol levels in a population of liberal arts college undergraduate students? Explore (NY) 2022; 19:283-289. [PMID: 35989236 DOI: 10.1016/j.explore.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 11/04/2022]
Abstract
The steroid hormone cortisol can be used to measure physiological stress in humans. The hypothalamic-pituitary-adrenal (HPA) axis synthesizes cortisol, and a negative feedback cycle regulates cortisol depending on an individual's stress level and/or circadian rhythm. Chronic stress of college undergraduate students is associated with various adverse health effects, including anxiety and depression. Reports suggest that stress levels have risen dramatically in recent years, particularly among university students dealing with intense academic loads in addition to COVID-19 pandemic-related uncertainty. The increasing rate of mental illness on college campuses necessitates the study of mediators potentially capable of lowering stress, and thus cortisol levels. Research on mediation techniques and coping mechanisms have gained traction to address the concerning levels of stress, including the employment of human-animal interaction sessions on college campuses. In this study, human-canine interaction as a stress mediation strategy for undergraduate students was investigated. We measured salivary cortisol levels in 73 college undergraduate students during a 60-min interaction period with a dog to determine whether human-canine interactions are effective in lowering cortisol levels and potentially reducing chronic stress typical of undergraduate students. Our results indicate that a human-canine interaction for 60 min is an effective method for significantly reducing salivary cortisol and stress levels among undergraduate college students. These findings support the expansion of animal visitation programs on college campuses to help students manage stress.
Collapse
Affiliation(s)
| | - Luke Calderaro
- Department of Biology, Colgate University, Hamilton, NY 13346, USA
| | - Sophia Clark
- Department of Biology, Colgate University, Hamilton, NY 13346, USA
| | - David Elacqua
- Department of Biology, Colgate University, Hamilton, NY 13346, USA
| | - Emily Hazen
- Department of Biology, Colgate University, Hamilton, NY 13346, USA
| | - Vanessa Lam
- Department of Biology, Colgate University, Hamilton, NY 13346, USA
| | | |
Collapse
|
37
|
Ouanes S, Clark C, Richiardi J, Maréchal B, Lewczuk P, Kornhuber J, Kirschbaum C, Popp J. Cerebrospinal Fluid Cortisol and Dehydroepiandrosterone Sulfate, Alzheimer’s Disease Pathology, and Cognitive Decline. Front Aging Neurosci 2022; 14:892754. [PMID: 35875796 PMCID: PMC9301040 DOI: 10.3389/fnagi.2022.892754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/22/2022] [Indexed: 12/31/2022] Open
Abstract
Introduction Elevated cortisol levels have been reported in Alzheimer’s disease (AD) and may accelerate the development of brain pathology and cognitive decline. Dehydroepiandrosterone sulfate (DHEAS) has anti-glucocorticoid effects and it may be involved in the AD pathophysiology. Objectives To investigate associations of cerebrospinal fluid (CSF) cortisol and DHEAS levels with (1) cognitive performance at baseline; (2) CSF biomarkers of amyloid pathology (as assessed by CSF Aβ levels), neuronal injury (as assessed by CSF tau), and tau hyperphosphorylation (as assessed by CSF p-tau); (3) regional brain volumes; and (4) clinical disease progression. Materials and Methods Individuals between 49 and 88 years (n = 145) with mild cognitive impairment or dementia or with normal cognition were included. Clinical scores, AD biomarkers, brain MRI volumetry along with CSF cortisol and DHEAS were obtained at baseline. Cognitive and functional performance was re-assessed at 18 and 36 months from baseline. We also assessed the following covariates: apolipoprotein E (APOE) genotype, BMI, and education. We used linear regression and mixed models to address associations of interest. Results Higher CSF cortisol was associated with poorer global cognitive performance and higher disease severity at baseline. Cortisol and cortisol/DHEAS ratio were positively associated with tau and p-tau CSF levels, and negatively associated with the amygdala and insula volumes at baseline. Higher CSF cortisol predicted more pronounced cognitive decline and clinical disease progression over 36 months. Higher CSF DHEAS predicted more pronounced disease progression over 36 months. Conclusion Increased cortisol in the CNS is associated with tau pathology and neurodegeneration, and with decreased insula and amygdala volume. Both CSF cortisol and DHEAS levels predict faster clinical disease progression. These results have implications for the identification of patients at risk of rapid decline as well as for the development of interventions targeting both neurodegeneration and clinical manifestations of AD.
Collapse
Affiliation(s)
- Sami Ouanes
- Service of Old Age Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Department of Psychiatry, Hamad Medical Corporation, Doha, Qatar
| | - Christopher Clark
- Centre for Gerontopsychiatric Medicine, Geriatric Psychiatry, University Hospital of Psychiatry Zürich, Zurich, Switzerland
| | - Jonas Richiardi
- Department of Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Bénédicte Maréchal
- Department of Radiology, Lausanne University Hospital, Lausanne, Switzerland
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Clemens Kirschbaum
- Chair of Biopsychology, Technische Universität Dresden, Andreas-Schubert-Bau, Dresden, Germany
| | - Julius Popp
- Service of Old Age Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Centre for Gerontopsychiatric Medicine, Geriatric Psychiatry, University Hospital of Psychiatry Zürich, Zurich, Switzerland
- *Correspondence: Julius Popp,
| |
Collapse
|
38
|
Dromard Y, Arango-Lievano M, Borie A, Dedin M, Fontanaud P, Torrent J, Garabedian MJ, Ginsberg SD, Jeanneteau F. Loss of glucocorticoid receptor phosphorylation contributes to cognitive and neurocentric damages of the amyloid-β pathway. Acta Neuropathol Commun 2022; 10:91. [PMID: 35733193 PMCID: PMC9219215 DOI: 10.1186/s40478-022-01396-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 12/22/2022] Open
Abstract
Aberrant cortisol and activation of the glucocorticoid receptor (GR) play an essential role in age-related progression of Alzheimer's disease (AD). However, the GR pathways required for influencing the pathobiology of AD dementia remain unknown. To address this, we studied an early phase of AD-like progression in the well-established APP/PS1 mouse model combined with targeted mutations in the BDNF-dependent GR phosphorylation sites (serines 134/267) using molecular, behavioral and neuroimaging approaches. We found that disrupting GR phosphorylation (S134A/S267A) in mice exacerbated the deleterious effects of the APP/PS1 genotype on mortality, neuroplasticity and cognition, without affecting either amyloid-β deposition or vascular pathology. The dynamics, maturation and retention of task-induced new dendritic spines of cortical excitatory neurons required GR phosphorylation at the BDNF-dependent sites that amyloid-β compromised. Parallel studies in postmortem human prefrontal cortex revealed AD subjects had downregulated BDNF signaling and concomitant upregulated cortisol pathway activation, which correlated with cognitive decline. These results provide key evidence that the loss of neurotrophin-mediated GR phosphorylation pathway promotes the detrimental effects of the brain cortisol response that contributes to the onset and/or progression of AD dementia. These findings have important translational implications as they provide a novel approach to treating AD dementia by identifying drugs that increase GR phosphorylation selectively at the neurotrophic sites to improve memory and cognition.
Collapse
Affiliation(s)
- Yann Dromard
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
| | - Margarita Arango-Lievano
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
| | - Amelie Borie
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
| | - Maheva Dedin
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
| | - Pierre Fontanaud
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France
- Imagerie du Petit Animal de Montpellier, 34090, Montpellier, France
| | - Joan Torrent
- Institut de Neuroscience de Montpellier, INSERM, 34090, Montpellier, France
| | - Michael J Garabedian
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Stephen D Ginsberg
- Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Departments of Psychiatry, Neuroscience & Physiology, NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Freddy Jeanneteau
- Institut de Génomiqueénomique Fonctionnelle, Université de Montpellier, INSERM, CNRS, 34090, Montpellier, France.
| |
Collapse
|
39
|
Nucara A, Ripanti F, Sennato S, Nisini G, De Santis E, Sefat M, Carbonaro M, Mango D, Minicozzi V, Carbone M. Influence of Cortisol on the Fibril Formation Kinetics of Aβ42 Peptide: A Multi-Technical Approach. Int J Mol Sci 2022; 23:ijms23116007. [PMID: 35682687 PMCID: PMC9180743 DOI: 10.3390/ijms23116007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 11/24/2022] Open
Abstract
Amyloid-β peptide (Aβ) aggregates are known to be correlated with pathological neurodegenerative diseases. The fibril formation process of such peptides in solution is influenced by several factors, such as the ionic strength of the buffer, concentration, pH, and presence of other molecules, just to mention a few. In this paper, we report a detailed analysis of in vitro Aβ42 fibril formation in the presence of cortisol at different relative concentrations. The thioflavin T fluorescence assay allowed us to monitor the fibril formation kinetics, while a morphological characterization of the aggregates was obtained by atomic force microscopy. Moreover, infrared absorption spectroscopy was exploited to investigate the secondary structure changes along the fibril formation path. Molecular dynamics calculations allowed us to understand the intermolecular interactions with cortisol. The combined results demonstrated the influence of cortisol on the fibril formation process: indeed, at cortisol-Aβ42 concentration ratio (ρ) close to 0.1 a faster organization of Aβ42 fragments into fibrils is promoted, while for ρ = 1 the formation of fibrils is completely inhibited.
Collapse
Affiliation(s)
- Alessandro Nucara
- Department of Physics, Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy;
- Correspondence: (A.N.); (F.R.)
| | - Francesca Ripanti
- Department of Physics and Geology, University of Perugia, Via Alessandro Pascoli, 06123 Perugia, Italy
- Correspondence: (A.N.); (F.R.)
| | - Simona Sennato
- CNR-ISC Sede Sapienza, Department of Physics, Sapienza University, P.le A. Moro 5, 00185 Rome, Italy;
| | - Giacomo Nisini
- Department of Physics, Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy;
| | - Emiliano De Santis
- Department of Physics and Astronomy and Department of Chemistry-BMC, Uppsala University, Husargatan 3, 752 37 Uppsala, Sweden;
| | - Mahta Sefat
- School of Pharmacy, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (M.S.); (D.M.)
| | - Marina Carbonaro
- Council for Agricultural Research and Economics (CREA), Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy;
| | - Dalila Mango
- School of Pharmacy, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (M.S.); (D.M.)
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, 00161 Rome, Italy
| | - Velia Minicozzi
- Department of Physics and INFN, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133 Rome, Italy;
| | - Marilena Carbone
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy;
| |
Collapse
|
40
|
Hoyt KR, Obrietan K. Circadian clocks, cognition, and Alzheimer's disease: synaptic mechanisms, signaling effectors, and chronotherapeutics. Mol Neurodegener 2022; 17:35. [PMID: 35525980 PMCID: PMC9078023 DOI: 10.1186/s13024-022-00537-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/08/2022] [Indexed: 12/20/2022] Open
Abstract
Modulation of basic biochemical and physiological processes by the circadian timing system is now recognized as a fundamental feature of all mammalian organ systems. Within the central nervous system, these clock-modulating effects are reflected in some of the most complex behavioral states including learning, memory, and mood. How the clock shapes these behavioral processes is only now beginning to be realized. In this review we describe recent findings regarding the complex set of cellular signaling events, including kinase pathways, gene networks, and synaptic circuits that are under the influence of the clock timing system and how this, in turn, shapes cognitive capacity over the circadian cycle. Further, we discuss the functional roles of the master circadian clock located in the suprachiasmatic nucleus, and peripheral oscillator populations within cortical and limbic circuits, in the gating of synaptic plasticity and memory over the circadian cycle. These findings are then used as the basis to discuss the connection between clock dysregulation and cognitive impairments resulting from Alzheimer's disease (AD). In addition, we discuss the conceptually novel idea that in AD, there is a selective disruption of circadian timing within cortical and limbic circuits, and that it is the disruption/desynchronization of these regions from the phase-entraining effects of the SCN that underlies aspects of the early- and mid-stage cognitive deficits in AD. Further, we discuss the prospect that the disruption of circadian timing in AD could produce a self-reinforcing feedback loop, where disruption of timing accelerates AD pathogenesis (e.g., amyloid deposition, oxidative stress and cell death) that in turn leads to a further disruption of the circadian timing system. Lastly, we address potential therapeutic approaches that could be used to strengthen cellular timing networks and, in turn, how these approaches could be used to improve cognitive capacity in Alzheimer's patients.
Collapse
Affiliation(s)
- Kari R Hoyt
- Division of Pharmaceutics and Pharmacology, Ohio State University, 412 Riffe Building, 12th Ave, Columbus, OH, 43210, USA.
| | - Karl Obrietan
- Department of Neuroscience, Ohio State University, Graves Hall, 333 W. 10th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
41
|
Montoliu T, Pulopulos MM, Puig-Pérez S, Hidalgo V, Salvador A. Mediation of perceived stress and cortisol in the association between neuroticism and global cognition in older adults: A longitudinal study. Stress Health 2022; 38:290-303. [PMID: 34363312 DOI: 10.1002/smi.3088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 11/06/2022]
Abstract
Neuroticism has been associated with a greater dementia risk, but its association with cognitive decline in healthy older adults remains unclear. Stress has been proposed as one of the mechanisms that could explain this relationship. Our aim was to analyse, in healthy older people, the mediating role of perceived stress and the Hypothalamic-Pituitary-Adrenal (HPA) axis in the association between neuroticism and global cognition. At Waves 1 and 2 (4-year follow-up), 87 older people (49.4% women; M age = 65.08, SD = 4.54 at Wave 1) completed a neuropsychological battery and the Perceived Stress Scale (PSS), and provided saliva samples on two (Wave 1) and three (Wave 2) consecutive days to measure the wake-to-bed slope. In Wave 2, neuroticism was assessed with the NEO-Five-Factor Inventory. PSS, but not the wake-to-bed slope, mediated the negative associations between neuroticism and global cognition (Waves 1, 2 and change). Regarding gender differences, PSS (Waves 1, 2 and change) and the wake-to-bed slope (Wave 2 and change) mediated these associations in men. Our results suggest that perceived stress and HPA-axis dysregulation could act as mechanisms underlying the association between neuroticism and cognitive functioning and decline, at least in older men.
Collapse
Affiliation(s)
- Teresa Montoliu
- Laboratory of Social Cognitive Neuroscience, Psychobiology-IDOCAL, Faculty of Psychology, University of Valencia, Valencia, Spain
| | - Matías M Pulopulos
- IIS Aragón, Department of Psychology and Sociology, Area of Psychobiology, University of Zaragoza, Teruel, Spain
| | - Sara Puig-Pérez
- Research Group of Psychology and Quality of Life, Valencian International University, Valencia, Spain
| | - Vanesa Hidalgo
- Laboratory of Social Cognitive Neuroscience, Psychobiology-IDOCAL, Faculty of Psychology, University of Valencia, Valencia, Spain.,IIS Aragón, Department of Psychology and Sociology, Area of Psychobiology, University of Zaragoza, Teruel, Spain
| | - Alicia Salvador
- Laboratory of Social Cognitive Neuroscience, Psychobiology-IDOCAL, Faculty of Psychology, University of Valencia, Valencia, Spain
| |
Collapse
|
42
|
Wu-Chung EL, Leal SL, Denny BT, Cheng SL, Fagundes CP. Spousal caregiving, widowhood, and cognition: A systematic review and a biopsychosocial framework for understanding the relationship between interpersonal losses and dementia risk in older adulthood. Neurosci Biobehav Rev 2022; 134:104487. [PMID: 34971701 PMCID: PMC8925984 DOI: 10.1016/j.neubiorev.2021.12.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/23/2021] [Accepted: 12/06/2021] [Indexed: 01/18/2023]
Abstract
Accumulating research suggests that stressful life events, especially those that threaten close intimate bonds, are associated with an increased risk of dementia. Grieving the loss of a spouse, whether in the form of caregiving or after the death, ranks among 'life's most significant stressors', evoking intense psychological and physiological distress. Despite numerous studies reporting elevated dementia risk or poorer cognition among spousal caregivers and widow(er)s compared to controls, no review has summarized findings across cognitive outcomes (i.e., dementia incidence, cognitive impairment rates, cognitive performance) or proposed a theoretical model for understanding the links between partner loss and abnormal cognitive decline. The current systematic review summarizes findings across 64 empirical studies. Overall, both cross-sectional and longitudinal studies revealed an adverse association between partner loss and cognitive outcomes. In turn, we propose a biopsychosocial model of cognitive decline that explains how caregiving and bereavement may position some to develop cognitive impairment or Alzheimer's disease and related dementias. More longitudinal studies that focus on the biopsychosocial context of caregivers and widow(er)s are needed.
Collapse
Affiliation(s)
- E Lydia Wu-Chung
- Department of Psychological Sciences, Rice University, Houston, TX, United States.
| | - Stephanie L Leal
- Department of Psychological Sciences, Rice University, Houston, TX, United States
| | - Bryan T Denny
- Department of Psychological Sciences, Rice University, Houston, TX, United States
| | - Samantha L Cheng
- Department of Psychological Sciences, Rice University, Houston, TX, United States
| | - Christopher P Fagundes
- Department of Psychological Sciences, Rice University, Houston, TX, United States; Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
43
|
Ferguson LA, Leal SL. Interactions of Emotion and Memory in the Aging Brain: Neural and Psychological Correlates. Curr Behav Neurosci Rep 2022. [DOI: 10.1007/s40473-021-00245-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
A Growing Link between Circadian Rhythms, Type 2 Diabetes Mellitus and Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23010504. [PMID: 35008933 PMCID: PMC8745289 DOI: 10.3390/ijms23010504] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) patients are at a higher risk of developing Alzheimer’s disease (AD). Mounting evidence suggests the emerging important role of circadian rhythms in many diseases. Circadian rhythm disruption is considered to contribute to both T2DM and AD. Here, we review the relationship among circadian rhythm disruption, T2DM and AD, and suggest that the occurrence and progression of T2DM and AD may in part be associated with circadian disruption. Then, we summarize the promising therapeutic strategies targeting circadian dysfunction for T2DM and AD, including pharmacological treatment such as melatonin, orexin, and circadian molecules, as well as non-pharmacological treatments like light therapy, feeding behavior, and exercise.
Collapse
|
45
|
Gerritsen L, Twait EL, Jonsson PV, Gudnason V, Launer LJ, Geerlings MI. Depression and Dementia: The Role of Cortisol and Vascular Brain Lesions. AGES-Reykjavik Study. J Alzheimers Dis 2022; 85:1677-1687. [PMID: 34958034 PMCID: PMC11044806 DOI: 10.3233/jad-215241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Late-life depression (LLD) is related to an increased risk of developing dementia; however, the biological mechanisms explaining this relationship remain unclear. OBJECTIVE To determine whether the relationship between LLD and dementia can be best explained by the glucocorticoid cascade or vascular hypothesis. METHODS Data are from 4,354 persons (mean age 76±5 years) without dementia at baseline from the AGES-Reykjavik Study. LLD was assessed with the MINI diagnostic interview (current and remitted major depressive disorder [MDD]) and the Geriatric Depression Scale-15. Morning and evening salivary cortisol were collected (glucocorticoid cascade hypothesis). White matter hyperintensities (WMH; vascular hypothesis) volume was assessed using 1.5T brain MRI. Using Cox proportional hazard models, we estimated the associations of LLD, cortisol levels, and WMH volume with incident all-cause dementia, AD, and non-AD dementia. RESULTS During 8.8±3.2 years of follow-up, 843 persons developed dementia, including 397 with AD. Current MDD was associated with an increased risk of developing all-cause dementia (HR = 2.17; 95% CI 1.66-2.67), with risks similar for AD and non-AD, while remitted MDD was not (HR = 1.02; 95% CI 0.55-1.49). Depressive symptoms were also associated with increased risk of dementia, in particular non-AD dementias. Higher levels of evening cortisol increased risk of dementia, but this was independent of MDD. WMH partially explained the relation between current MDD and dementia risk but remained increased (HR = 1.71; 95% CI 1.34-2.08). CONCLUSION The current study highlights the importance of LLD in developing dementia. However, neither the glucocorticoid cascade nor the vascular hypotheses fully explained the relation between depression and dementia.
Collapse
Affiliation(s)
- Lotte Gerritsen
- Department of Psychology, Utrecht University, Utrecht, the Netherlands
| | - Emma L. Twait
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Palmi V. Jonsson
- Department of Geriatrics, Landspitali University Hospital, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Vilmundur Gudnason
- Department of Psychology, Utrecht University, Utrecht, the Netherlands
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Lenore J. Launer
- National Institute on Aging, Laboratory for Epidemiology and Population Sciences, Baltimore, MD, USA
| | - Mirjam I. Geerlings
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
- National Institute on Aging, Laboratory for Epidemiology and Population Sciences, Baltimore, MD, USA
| |
Collapse
|
46
|
Enduring glucocorticoid-evoked exacerbation of synaptic plasticity disruption in male rats modelling early Alzheimer's disease amyloidosis. Neuropsychopharmacology 2021; 46:2170-2179. [PMID: 34188184 PMCID: PMC8505492 DOI: 10.1038/s41386-021-01056-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/12/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
Synaptic dysfunction is a likely proximate cause of subtle cognitive impairment in early Alzheimer's disease. Soluble oligomers are the most synaptotoxic forms of amyloid ß-protein (Aß) and mediate synaptic plasticity disruption in Alzheimer's disease amyloidosis. Because the presence and extent of cortisol excess in prodromal Alzheimer's disease predicts the onset of cognitive symptoms we hypothesised that corticosteroids would exacerbate the inhibition of hippocampal synaptic long-term potentiation in a rat model of Alzheimer's disease amyloidosis. In a longitudinal experimental design using freely behaving pre-plaque McGill-R-Thy1-APP male rats, three injections of corticosterone or the glucocorticoid methylprednisolone profoundly disrupted long-term potentiation induced by strong conditioning stimulation for at least 2 months. The same treatments had a transient or no detectible detrimental effect on synaptic plasticity in wild-type littermates. Moreover, corticosterone-mediated cognitive dysfunction, as assessed in a novel object recognition test, was more persistent in the transgenic animals. Evidence for the involvement of pro-inflammatory mechanisms was provided by the ability of the selective the NOD-leucine rich repeat and pyrin containing protein 3 (NLRP3) inflammasome inhibitor Mcc950 to reverse the synaptic plasticity deficit in corticosterone-treated transgenic animals. The marked prolongation of the synaptic plasticity disrupting effects of brief corticosteroid excess substantiates a causal role for hypothalamic-pituitary-adrenal axis dysregulation in early Alzheimer's disease.
Collapse
|
47
|
Faraji J, Metz GAS. Aging, Social Distancing, and COVID-19 Risk: Who is more Vulnerable and Why? Aging Dis 2021; 12:1624-1643. [PMID: 34631211 PMCID: PMC8460299 DOI: 10.14336/ad.2021.0319] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/19/2021] [Indexed: 01/08/2023] Open
Abstract
Perceived social support represents an important predictor of healthy aging. The global COVID-19 pandemic has dramatically changed the face of social relationships and revealed elderly to be particularly vulnerable to the effects of social isolation. Social distancing may represent a double-edged sword for older adults, protecting them against COVID-19 infection while also sacrificing personal interaction and attention at a critical time. Here, we consider the moderating role of social relationships as a potential influence on stress resilience, allostatic load, and vulnerability to infection and adverse health outcomes in the elderly population. Understanding the mechanisms how social support enhances resilience to stress and promotes mental and physical health into old age will enable new preventive strategies. Targeted social interventions may provide effective relief from the impact of COVID-19-related isolation and loneliness. In this regard, a pandemic may also offer a window of opportunity for raising awareness and mobilizing resources for new strategies that help build resilience in our aging population and future generations.
Collapse
Affiliation(s)
- Jamshid Faraji
- 1Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada.,2Faculty of Nursing & Midwifery, Golestan University of Medical Sciences, Gorgan, Iran
| | - Gerlinde A S Metz
- 1Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
48
|
Saelzler UG, Verhaeghen P, Panizzon MS, Moffat SD. Intact circadian rhythm despite cortisol hypersecretion in Alzheimer's disease: A meta-analysis. Psychoneuroendocrinology 2021; 132:105367. [PMID: 34340133 DOI: 10.1016/j.psyneuen.2021.105367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/15/2021] [Accepted: 07/22/2021] [Indexed: 12/26/2022]
Abstract
Hypersecretion of the glucocorticoid steroid hormone cortisol by individuals with Alzheimer's disease (AD) has been suspected for several decades, during which time dozens of examinations of this phenomenon have been conducted and published. The goals of this investigation were to summarize this sizeable body of literature, test whether participant and methodological characteristics modify the magnitude of the AD-associated basal cortisol hypersecretion, and examine whether cortisol circadian rhythmicity is maintained among individuals with AD. To this end, the present meta-analysis and systematic review examined over 300 comparisons of indices of basal HPA-axis functioning between individuals with AD and cognitively normal older adults. AD was associated with basal cortisol elevations (g = 0.45) but the magnitude of the effect was not systematically impacted by any of the participant characteristics considered or the time-of-day of the cortisol sampling. Further, there was no evidence of group differences among direct indices of circadian rhythmicity such as the cortisol awakening response or the diurnal cortisol slope. These results suggest that basal hypersecretion of cortisol, but not circadian dysrhythmia, is characteristic of individuals with AD. Mechanistically, the observed hypersecretion is consistent with the theorized AD-driven deterioration of the hippocampus and subsequent reduction in hypothalamic-pituitary-adrenal axis inhibition. Further investigation is warranted to elucidate the role and timing of cortisol elevations in the progression of AD.
Collapse
Affiliation(s)
- Ursula G Saelzler
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr. La Jolla, San Diego, CA 92093, USA.
| | - Paul Verhaeghen
- Department of Psychology, Georgia Institute of Technology, 648 Cherry St. NW, Atlanta GA 30313, USA.
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr. La Jolla, San Diego, CA 92093, USA.
| | - Scott D Moffat
- Department of Psychology, Georgia Institute of Technology, 648 Cherry St. NW, Atlanta GA 30313, USA.
| |
Collapse
|
49
|
Ancelin ML, Norton J, Scali J, Ritchie K, Chaudieu I, Ryan J. A Prospective Study of Diurnal Cortisol and Incident Dementia in Community-Dwelling Older Adults. J Alzheimers Dis 2021; 82:899-904. [PMID: 34120906 DOI: 10.3233/jad-210389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Diurnal salivary cortisol was measured in 334 older adults without dementia, at four times on two separate days, under quiet and stressful conditions. In multivariate Cox proportional hazard models, higher global diurnal cortisol secretion was associated with incident dementia (HR = 1.09 [1.02-1.15] per one-unit increase in cortisol measure, p = 0.007) and Alzheimer's disease (HR = 1.12 [1.04-1.21], p = 0.003) over a mean (SD) of 8.1 (4.0) years, independent of potential confounders and stressful conditions. Individuals with incident dementia had a slower rate of cortisol elimination under non-stressful conditions, reflected by higher cortisol levels in the evening, and an abnormal response to stress (blunted evening stress response).
Collapse
Affiliation(s)
| | | | | | - Karen Ritchie
- INM, Univ Montpellier, INSERM, Montpellier, France.,Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | | | - Joanne Ryan
- Biological Neuropsychiatry Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
50
|
Milligan Armstrong A, Porter T, Quek H, White A, Haynes J, Jackaman C, Villemagne V, Munyard K, Laws SM, Verdile G, Groth D. Chronic stress and Alzheimer's disease: the interplay between the hypothalamic-pituitary-adrenal axis, genetics and microglia. Biol Rev Camb Philos Soc 2021; 96:2209-2228. [PMID: 34159699 DOI: 10.1111/brv.12750] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/21/2022]
Abstract
Chronic psychosocial stress is increasingly being recognised as a risk factor for sporadic Alzheimer's disease (AD). The hypothalamic-pituitary-adrenal axis (HPA axis) is the major stress response pathway in the body and tightly regulates the production of cortisol, a glucocorticoid hormone. Dysregulation of the HPA axis and increased levels of cortisol are commonly found in AD patients and make a major contribution to the disease process. The underlying mechanisms remain poorly understood. In addition, within the general population there are interindividual differences in sensitivities to glucocorticoid and stress responses, which are thought to be due to a combination of genetic and environmental factors. These differences could ultimately impact an individuals' risk of AD. The purpose of this review is first to summarise the literature describing environmental and genetic factors that can impact an individual's HPA axis reactivity and function and ultimately AD risk. Secondly, we propose a mechanism by which genetic factors that influence HPA axis reactivity may also impact inflammation, a key driver of neurodegeneration. We hypothesize that these factors can mediate glucocorticoid priming of the immune cells of the brain, microglia, to become pro-inflammatory and promote a neurotoxic environment resulting in neurodegeneration. Understanding the underlying molecular mechanisms and identifying these genetic factors has implications for evaluating stress-related risk/progression to neurodegeneration, informing the success of interventions based on stress management and potential risks associated with the common use of glucocorticoids.
Collapse
Affiliation(s)
- Ayeisha Milligan Armstrong
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Tenielle Porter
- Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
| | - Hazel Quek
- QIMR Berghofer Medical Institute, 300 Herston Rd, Herston, QLD, Australia
| | - Anthony White
- QIMR Berghofer Medical Institute, 300 Herston Rd, Herston, QLD, Australia
| | - John Haynes
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Connie Jackaman
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Victor Villemagne
- School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Kylie Munyard
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia
| | - Simon M Laws
- Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
| | - Giuseppe Verdile
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia.,School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, WA, 6027, Australia
| | - David Groth
- Curtin Health Innovation Research Institute, Curtin University, Kent St, Bentley, WA, 6102, Australia.,Curtin Medical School, Curtin University, Kent St, Bentley, WA, 6102, Australia
| |
Collapse
|