1
|
Scarpetta V, Ho KH, Trapp M, Patrizi A. Choroid plexus: Insights from distinct epithelial cellular components. Curr Opin Neurobiol 2025; 93:103028. [PMID: 40267629 DOI: 10.1016/j.conb.2025.103028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/25/2025]
Abstract
The choroid plexus (ChP) serves as a vital interface between blood and cerebrospinal fluid (CSF), playing a pivotal role in central nervous system (CNS) development and communication with the body. This review mainly summarizes how the ChP epithelial cells respond to physiological and pathological stimuli, emphasizing the role of distinct organelles and key molecular signaling pathways. Additionally, we discuss the roles of ChP cilia, an evolutionary conserved organelle whose function is still under investigation. Understanding these processes is essential for elucidating how ChP function modulates intrinsic and extrinsic stimuli, which are crucial for maintaining CNS and body homeostasis.
Collapse
Affiliation(s)
- Valentina Scarpetta
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neurosciences "Rita Levi Montalcini", University of Turin, Turin 10126, Italy
| | - Kim Hoa Ho
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg 69120, Germany
| | - Marleen Trapp
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Annarita Patrizi
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Interdisciplinary Center for Neuroscience, Heidelberg University, Heidelberg 69120, Germany.
| |
Collapse
|
2
|
Tack RWP, Amboni C, van Nuijs D, Pekna M, Vergouwen MDI, Rinkel GJE, Hol EM. Inflammation, Anti-inflammatory Interventions, and Post-stroke Cognitive Impairment: a Systematic Review and Meta-analysis of Human and Animal Studies. Transl Stroke Res 2025; 16:535-546. [PMID: 38012509 PMCID: PMC11976800 DOI: 10.1007/s12975-023-01218-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023]
Abstract
The pathophysiology and treatment of post-stroke cognitive impairment (PSCI) are not clear. Stroke triggers an inflammatory response, which might affect synapse function and cognitive status. We performed a systematic review and meta-analysis to assess whether patients with PSCI have increased levels of inflammatory markers and whether anti-inflammatory interventions in animals decrease PSCI. We systematically searched PubMed, EMBASE, and PsychInfo for studies on stroke. For human studies, we determined the standardized mean difference (SMD) on the association between PSCI and markers of inflammation. For animal studies, we determined the SMD of post-stroke cognitive outcome after an anti-inflammatory intervention. Interventions were grouped based on proposed mechanism of action. In patients, the SMD of inflammatory markers for those with versus those without PSCI was 0.46 (95% CI 0.18; 0.76; I2 = 92%), and the correlation coefficient between level of inflammation and cognitive scores was - 0.25 (95% CI - 0.34; - 0.16; I2 = 75%). In animals, the SMD of cognition for those treated with versus those without anti-inflammatory interventions was 1.43 (95% CI 1.12; 1.74; I2 = 83%). The largest effect sizes in treated animals were for complement inhibition (SMD = 1.94 (95% CI 1.50; 2.37), I2 = 51%) and fingolimod (SMD = 2.1 (95% CI 0.75; 3.47), I2 = 81%). Inflammation is increased in stroke survivors with cognitive impairment and is negatively correlated with cognitive functioning. Anti-inflammatory interventions seem to improve cognitive functioning in animals. Complement inhibition and fingolimod are promising therapies on reducing PSCI.
Collapse
Affiliation(s)
- Reinier W P Tack
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| | - Claudia Amboni
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Danny van Nuijs
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Marcela Pekna
- Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mervyn D I Vergouwen
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Gabriel J E Rinkel
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Taha B, McGovern R, Lam C. A synthesized view of the CSF-blood barrier and its surgical implications for aging disorders. Front Aging Neurosci 2025; 16:1492449. [PMID: 39981073 PMCID: PMC11841429 DOI: 10.3389/fnagi.2024.1492449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/30/2024] [Indexed: 02/22/2025] Open
Abstract
In this review, we explore the mechanisms of the blood-cerebrospinal fluid (CSF) barrier and CSF transport. We briefly review the mathematical framework for CSF transport as described by a set of well-studied partial differential equations. Moreover, we describe the major contributors of CSF flow through both diffusive and convective forces beginning at the molecular level and extending into macroscopic clinical observations. In addition, we review neurosurgical perspectives in understanding CSF outflow pathways. Finally, we discuss the implications of flow dysregulation in the context of neurodegenerative diseases and discuss the rising role of perivascular drainage pathways including glymphatics.
Collapse
Affiliation(s)
- Birra Taha
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Robert McGovern
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Cornelius Lam
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
- Minneapolis VA Health Care System, Veterans Health Administration, United States Department of Veterans Affairs, Minneapolis, MN, United States
| |
Collapse
|
4
|
Morató X, Puerta R, Cano A, Orellana A, de Rojas I, Capdevila M, Montrreal L, Rosende-Roca M, García-González P, Olivé C, García-Gutiérrez F, Blázquez J, Miguel A, Núñez-Llaves R, Pytel V, Alegret M, Fernández MV, Marquié M, Valero S, Cavazos JE, Mañes S, Boada M, Cabrera-Socorro A, Ruiz A. Associations of plasma SMOC1 and soluble IL6RA levels with the progression from mild cognitive impairment to dementia. Brain Behav Immun Health 2024; 42:100899. [PMID: 39640195 PMCID: PMC11617377 DOI: 10.1016/j.bbih.2024.100899] [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: 05/03/2024] [Revised: 08/12/2024] [Accepted: 10/27/2024] [Indexed: 12/07/2024] Open
Abstract
Despite the central role attributed to neuroinflammation in the etiology and pathobiology of Alzheimer's disease (AD), the direct link between levels of inflammatory mediators in blood and cerebrospinal fluid (CSF) compartments, as well as their potential implications for AD diagnosis and progression, remains inconclusive. Moreover, there is debate on whether inflammation has a protective or detrimental effect on disease onset and progression. Indeed, distinct immunological mechanisms may govern protective and damaging effects at early and late stages, respectively. This study aims to (i) identify inflammatory mediators demonstrating robust correlations between peripheral and central nervous system (CNS) compartments by means of plasma and CSF analysis, respectively, and (ii) assess their potential significance in the context of AD and disease progression from mild cognitive impairment (MCI) to dementia. To achieve this, we have examined the inflammatory profile of a well-defined subcohort comprising 485 individuals from the Ace Alzheimer Center Barcelona (ACE). Employing a hierarchical clustering approach, we thoroughly evaluated the intercompartmental correlations of 63 distinct inflammation mediators, quantified in paired CSF and plasma samples, using advanced SOMAscan technology. Of the array of mediators investigated, only six mediators (CRP, IL1RAP, ILRL1, IL6RA, PDGFRB, and YKL-40) exhibited robust correlations between the central and peripheral compartments (proximity scores <400). To strengthen the validity of our findings, these identified mediators were subsequently validated in a second subcohort of individuals from ACE (n = 873). The observed plasma correlations across the entire cohort consistently have a Spearman rho value above 0.51 (n = 1,360, p < 1.77E-93). Of the high CSF-plasma correlated proteins, only soluble IL6RA (sIL6RA) displayed a statistically significant association with the conversion from MCI to dementia. This association remained robust even after applying a stringent Bonferroni correction (Cox proportional hazard ratio [HR] = 1.936 per standard deviation; p = 0.0018). This association retained its significance when accounting for various factors, including CSF amyloid (Aβ42) and Thr181-phosphorylated tau (p-tau) levels, age, sex, baseline Mini-Mental State Examination (MMSE) score, and potential sampling biases identified through principal component analysis (PCA) modeling. Furthermore, our study confirmed the association of both plasma and CSF levels of SPARC-related modular calcium-binding protein 1 (SMOC1) with amyloid and tau accumulation, indicating their role as early surrogate biomarkers for AD pathology. Despite the lack of a statistically significant correlation between SMOC1 levels in CSF and plasma, both acted as independent biomarkers of disease progression (HR > 1.3, p < 0.002). In conclusion, our study unveils that sIL6RA and SMOC1 are associated with MCI progression. The absence of correlations among inflammatory mediators between the central and peripheral compartments appears to be a common pattern, with only a few intriguing exceptions.
Collapse
Affiliation(s)
- Xavier Morató
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Raquel Puerta
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Universitat de Barcelona (UB), Spain
| | - Amanda Cano
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Spain
| | - Adelina Orellana
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - María Capdevila
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Spain
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Maitée Rosende-Roca
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Claudia Olivé
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Josep Blázquez
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Andrea Miguel
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Raúl Núñez-Llaves
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - Montserrat Alegret
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Marta Marquié
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Enrique Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), 28049, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Agustín Ruiz
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
5
|
Taheri S, Prestopnik J, Rosenberg GA. Barriers of the CNS transfer rate dynamics in patients with vascular cognitive impairment and dementia. Front Aging Neurosci 2024; 16:1462302. [PMID: 39385834 PMCID: PMC11461252 DOI: 10.3389/fnagi.2024.1462302] [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: 07/09/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
Background Advances in in vivo MRI techniques enable cerebral barrier transfer rates (K trans ) measurement in patients with vascular cognitive impairment and dementia (VCID). However, a consensus has not been reached on the dynamic contribution and importance of cerebral barrier abnormalities to the differential diagnosis of dementia subtypes. Our goal was to investigate the dynamics of blood-brain barrier (BBB) and blood-CSF barrier (BCSFB) K trans in patients with VCID longitudinally and determine the effect of aging. Methods We studied subjects at two time points over two years; they were 65.5 years of age (SD = 15.94, M/F = 24/14) at the first visit. We studied 38 patients, 18 of whom had two visits. We calculated the BBB and BCSFB K trans with dynamic contrast-enhanced T1 MR, and we used 1H-MR spectroscopy to measure N-acetylaspartate (NAA) levels in the white matter as a marker of injury. In addition, we measured CSF levels of active-matrix metalloproteinase-3 (MMP3) as an inflammatory biomarker to aid in patient clustering. Results Longitudinal BBB measurements revealed variable dynamic behavior: after two years, the BBB K trans increased in 55% of patients and decreased in the remaining 45% unpredictably. We did not find a significant linear model of BBB K trans versus age for VCID. For healthy controls, the model was K trans = 0.0014 + 0.0002 × age, which was significant (p = 0.046). VCID patients showed a reduction in BCSFB K trans compared to healthy controls (p = 0.01). Combining NAA, CSF MMP3, and K trans in a clustering analysis separated patients into groups. Conclusion These results suggest that BBB K trans in VCID is dynamic and BCSFB K trans reduced by age. By combining inflammatory biomarkers with BBB K trans data, it is possible to separate VCID patients into distinct groups with different underlying pathologies.
Collapse
Affiliation(s)
- Saeid Taheri
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, FL, United States
- Center for Functional and Molecular Imaging, University of South Florida (USF) Heart Institute, Tampa, FL, United States
| | | | - Gary A. Rosenberg
- Center for Memory and Aging, Albuquerque, NM, United States
- Department of Neurology, Health Sciences Center, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
6
|
Yang Y, Tong M, de la Monte SM. Early-Stage Moderate Alcohol Feeding Dysregulates Insulin-Related Metabolic Hormone Expression in the Brain: Potential Links to Neurodegeneration Including Alzheimer's Disease. J Alzheimers Dis Rep 2024; 8:1211-1228. [PMID: 39247872 PMCID: PMC11380283 DOI: 10.3233/adr-240026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
Background Alzheimer's disease (AD), one of the most prevalent causes of dementia, is mainly sporadic in occurrence but driven by aging and other cofactors. Studies suggest that excessive alcohol consumption may increase AD risk. Objective Our study examined the degree to which short-term moderate ethanol exposure leads to molecular pathological changes of AD-type neurodegeneration. Methods Long Evans male and female rats were fed for 2 weeks with isocaloric liquid diets containing 24% or 0% caloric ethanol (n = 8/group). The frontal lobes were used to measure immunoreactivity to AD biomarkers, insulin-related endocrine metabolic molecules, and proinflammatory cytokines/chemokines by duplex or multiplex enzyme-linked immunosorbent assays (ELISAs). Results Ethanol significantly increased frontal lobe levels of phospho-tau, but reduced Aβ, ghrelin, glucagon, leptin, PAI, IL-2, and IFN-γ. Conclusions Short-term effects of chronic ethanol feeding produced neuroendocrine molecular pathologic changes reflective of metabolic dysregulation, together with abnormalities that likely contribute to impairments in neuroplasticity. The findings suggest that chronic alcohol consumption rapidly establishes a platform for impairments in energy metabolism that occur in both the early stages of AD and alcohol-related brain degeneration.
Collapse
Affiliation(s)
- Yiwen Yang
- Molecular Pharmacology, Physiology and Biotechnology Graduate Program, Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Suzanne M. de la Monte
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, the Providence VA Medical Center, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Departments of Neurology and Neurosurgery, Rhode Island Hospital, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
7
|
Kerl HU, Baazaoui H, Herrmann K, Adlung A, Ludwig NK, Hausner L, Frölich L, Schad L, Groden C, Mohamed SA. Sodium signal intensity of CSF using 1H-guided 23Na-MRI as a potential noninvasive biomarker in Alzheimer's disease. J Neuroimaging 2024; 34:619-626. [PMID: 38807265 DOI: 10.1111/jon.13216] [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] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND AND PURPOSE Alzheimer's disease (AD) is characterized by cognitive decline and mnestic deficits. The pathophysiology of AD is not fully understood, which renders the development of accurate tools for early diagnosis and effective therapies exceedingly difficult. In this study, we investigated the use of 23Na-MRI to measure the relative sodium signal intensities (rSSIs) in CSF in patients with AD and healthy controls. METHODS We prospectively recruited 11 patients with biomarker-diagnosed early-stage AD, as well as 12 cognitively healthy age-matched controls. All participants underwent 23Na-MRI to measure rSSI. Statistical analyses were performed to compare CSF sodium signal intensities between groups and to evaluate the specificity and sensitivity of the rSSI in the diagnosis of AD. RESULTS RSSIs in CSF were significantly higher in AD patients (mean = 68.6% ± 7.7%) compared to healthy controls (mean = 56.9% ± 5.5%) (p < .001). There was also a significant negative correlation between rSSI in CSF and hippocampus and amygdala volumes (r = -.54 and -.49, p < .05) as well as a positive correlation with total CSF volumes (r = .81, p < .05). Receiver operating characteristic analysis showed high diagnostic accuracy for rSSI in discriminating between AD patients and healthy controls (area under the curve = .94). CONCLUSION Our study provides evidence that rSSI in CSF is increased in AD patients in comparison to healthy controls. rSSI may serve as a potential marker for early detection and monitoring of disease progression. Larger, longitudinal studies are needed to confirm our findings and to investigate the association between rSSI in CSF and the severity of cognitive impairment.
Collapse
Affiliation(s)
- Hans-Ulrich Kerl
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hakim Baazaoui
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Katrin Herrmann
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anne Adlung
- Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Nadia K Ludwig
- Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Löwenstein Medical Technology, Karlsruhe, Germany
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Lothar Schad
- Department of Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Groden
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sherif A Mohamed
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
8
|
Niccolai E, Pedone M, Martinelli I, Nannini G, Baldi S, Simonini C, Di Gloria L, Zucchi E, Ramazzotti M, Spezia PG, Maggi F, Quaranta G, Masucci L, Bartolucci G, Stingo FC, Mandrioli J, Amedei A. Amyotrophic lateral sclerosis stratification: unveiling patterns with virome, inflammation, and metabolism molecules. J Neurol 2024; 271:4310-4325. [PMID: 38644373 PMCID: PMC11233352 DOI: 10.1007/s00415-024-12348-7] [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: 01/30/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/23/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is an untreatable and clinically heterogeneous condition primarily affecting motor neurons. The ongoing quest for reliable biomarkers that mirror the disease status and progression has led to investigations that extend beyond motor neurons' pathology, encompassing broader systemic factors such as metabolism, immunity, and the microbiome. Our study contributes to this effort by examining the potential role of microbiome-related components, including viral elements, such as torque tenovirus (TTV), and various inflammatory factors, in ALS. In our analysis of serum samples from 100 ALS patients and 34 healthy controls (HC), we evaluated 14 cytokines, TTV DNA load, and 18 free fatty acids (FFA). We found that the evaluated variables are effective in differentiating ALS patients from healthy controls. In addition, our research identifies four unique patient clusters, each characterized by distinct biological profiles. Intriguingly, no correlations were found with site of onset, sex, progression rate, phenotype, or C9ORF72 expansion. A remarkable aspect of our findings is the discovery of a gender-specific relationship between levels of 2-ethylhexanoic acid and patient survival. In addition to contributing to the growing body of evidence suggesting altered peripheral immune responses in ALS, our exploratory research underscores metabolic diversity challenging conventional clinical classifications. If our exploratory findings are validated by further research, they could significantly impact disease understanding and patient care customization. Identifying groups based on biological profiles might aid in clustering patients with varying responses to treatments.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Matteo Pedone
- Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Florence, Italy
| | - Ilaria Martinelli
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Cecilia Simonini
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Leandro Di Gloria
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Elisabetta Zucchi
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Matteo Ramazzotti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Pietro Giorgio Spezia
- Department of Translational Research, Retrovirus Center - University of Pisa, Pisa, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani - IRCCS, Rome, Italy
| | - Gianluca Quaranta
- Department of Laboratory and Infectious Sciences, A. Gemelli University Hospital IRCCS, Rome, Italy
| | - Luca Masucci
- Department of Laboratory and Infectious Sciences, A. Gemelli University Hospital IRCCS, Rome, Italy
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| | - Francesco Claudio Stingo
- Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Florence, Italy
| | - Jessica Mandrioli
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
9
|
McRae SA, Richards CM, Da Silva DE, Riar I, Yang SS, Zurfluh NE, Gibon J, Klegeris A. Pro-neuroinflammatory and neurotoxic potential of extracellular histones H1 and H3. Neurosci Res 2024; 204:34-45. [PMID: 38278218 DOI: 10.1016/j.neures.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/23/2023] [Accepted: 01/17/2024] [Indexed: 01/28/2024]
Abstract
Histones organize DNA within cellular nuclei, but they can be released from damaged cells. In peripheral tissues extracellular histones act as damage-associated molecular patterns (DAMPs) inducing pro-inflammatory activation of immune cells. Limited studies have considered DAMP-like activity of histones in the central nervous system (CNS); therefore, we studied the effects of extracellular histones on microglia, the CNS immunocytes, and on neuronal cells. Both the linker histone H1 and the core histone H3 induced pro-inflammatory activation of microglia-like cells by upregulating their secretion of NO and cytokines, including interferon-γ-inducible protein 10 (IP-10) and tumor necrosis factor-α (TNF). The selective inhibitors MMG-11 and TAK-242 were used to demonstrate involvement of toll-like receptors (TLR) 2 and 4, respectively, in H1-induced NO secretion by BV-2 microglia. H1, but not H3, downregulated the phagocytic activity of BV-2 microglia. H1 was also directly toxic to all neuronal cell types studied. We conclude that H1, and to a lesser extent H3, when released extracellularly, have the potential to act as a CNS DAMPs. Inhibition of the DAMP-like effects of extracellular histones on microglia and their neurotoxic activity represents a potential strategy for combating neurodegenerative diseases that are characterized by the adverse activation of microglia and neuronal death.
Collapse
Affiliation(s)
- Seamus A McRae
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Christy M Richards
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Dylan E Da Silva
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Ishvin Riar
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Sijie Shirley Yang
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Noah E Zurfluh
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Julien Gibon
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC V1V 1V7, Canada.
| |
Collapse
|
10
|
Lu Q, Yu A, Pu J, Chen D, Zhong Y, Bai D, Yang L. Post-stroke cognitive impairment: exploring molecular mechanisms and omics biomarkers for early identification and intervention. Front Mol Neurosci 2024; 17:1375973. [PMID: 38845616 PMCID: PMC11153683 DOI: 10.3389/fnmol.2024.1375973] [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: 01/24/2024] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Abstract
Post-stroke cognitive impairment (PSCI) is a major stroke consequence that has a severe impact on patients' quality of life and survival rate. For this reason, it is especially crucial to identify and intervene early in high-risk groups during the acute phase of stroke. Currently, there are no reliable and efficient techniques for the early diagnosis, appropriate evaluation, or prognostication of PSCI. Instead, plenty of biomarkers in stroke patients have progressively been linked to cognitive impairment in recent years. High-throughput omics techniques that generate large amounts of data and process it to a high quality have been used to screen and identify biomarkers of PSCI in order to investigate the molecular mechanisms of the disease. These techniques include metabolomics, which explores dynamic changes in the organism, gut microbiomics, which studies host-microbe interactions, genomics, which elucidates deeper disease mechanisms, transcriptomics and proteomics, which describe gene expression and regulation. We looked through electronic databases like PubMed, the Cochrane Library, Embase, Web of Science, and common databases for each omics to find biomarkers that might be connected to the pathophysiology of PSCI. As all, we found 34 studies: 14 in the field of metabolomics, 5 in the field of gut microbiomics, 5 in the field of genomics, 4 in the field of transcriptomics, and 7 in the field of proteomics. We discovered that neuroinflammation, oxidative stress, and atherosclerosis may be the primary causes of PSCI development, and that metabolomics may play a role in the molecular mechanisms of PSCI. In this study, we summarized the existing issues across omics technologies and discuss the latest discoveries of PSCI biomarkers in the context of omics, with the goal of investigating the molecular causes of post-stroke cognitive impairment. We also discuss the potential therapeutic utility of omics platforms for PSCI mechanisms, diagnosis, and intervention in order to promote the area's advancement towards precision PSCI treatment.
Collapse
Affiliation(s)
- Qiuyi Lu
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chonging, China
| | - Anqi Yu
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chonging, China
| | - Juncai Pu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chonging, China
| | - Dawei Chen
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chonging, China
| | - Yujie Zhong
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chonging, China
| | - Dingqun Bai
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chonging, China
| | - Lining Yang
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chonging, China
| |
Collapse
|
11
|
Shen M, Zhang L, Chen C, Wei X, Ma Y, Ma Y. Investigating the causal relationship between immune cell and Alzheimer's disease: a mendelian randomization analysis. BMC Neurol 2024; 24:98. [PMID: 38500057 PMCID: PMC10946133 DOI: 10.1186/s12883-024-03599-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/12/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Complex interactions between the immune system and the brain may affect neural development, survival, and function, with etiological and therapeutic implications for neurodegenerative diseases. However, previous studies investigating the association between immune inflammation and Alzheimer's disease (AD) have yielded inconsistent results. METHODS We applied Mendelian randomization (MR) to examine the causal relationship between immune cell traits and AD risk using genetic variants as instrumental variables. MR is an epidemiological study design based on genetic information that reduces the effects of confounding and reverse causation. We analyzed the causal associations between 731 immune cell traits and AD risk based on publicly available genetic data. RESULTS We observed that 5 immune cell traits conferred protection against AD, while 7 immune cell traits increased the risk of AD. These immune cell traits mainly involved T cell regulation, monocyte activation and B cell differentiation. Our findings suggest that immune regulation may influence the development of AD and provide new insights into potential targets for AD prevention and treatment. We also conducted various sensitivity analyses to test the validity and robustness of our results, which revealed no evidence of pleiotropy or heterogeneity. CONCLUSION Our research shows that immune regulation is important for AD and provides new information on potential targets for AD prevention and treatment. However, this study has limitations, including the possibility of reverse causality, lack of validation in independent cohorts, and potential confounding by population stratification. Further research is needed to validate and amplify these results and to elucidate the potential mechanisms of the immune cell-AD association.
Collapse
Affiliation(s)
- Min Shen
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Linlin Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chen Chen
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Xiaocen Wei
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yuning Ma
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China.
| | - Yuxia Ma
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China.
| |
Collapse
|
12
|
Wang L, Sha H, He X, Xie Y, Deng J, Chen J, Li G, Yang J. Neonatal IL-4 Over-Exposure is Accompanied by Macrophage Accumulation in Dura Mater After Instant Anti-inflammatory Cytokine Response in CSF. Cell Mol Neurobiol 2024; 44:18. [PMID: 38315435 PMCID: PMC10844484 DOI: 10.1007/s10571-023-01451-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 12/28/2023] [Indexed: 02/07/2024]
Abstract
Multiple studies have shown that clinical events resulting into neonatal IL-4 over-exposure, such as asthma in early life and food allergy, were associated with brain damage and that the neuroinflammation induced by them might lead to cognitive impairments, anxiety-/depressive-like behaviors. IL-4 is the most major elevated cytokine in periphery when these clinical events occur and peripheral IL-4 level positively correlates with the severity of those events. Our previous studies have verified that neonatal IL-4 over-exposure induced a delayed neuroinflammatory damage in rodents, which might have adverse implications for brain development and cognition. Neuroinflammation in brain parenchyma is often accompanied by changes in CSF cytokines levels. However, whether the cytokines levels in CSF change after neonatal IL-4 over-exposure is unknown. Here, we found a delayed pro-inflammatory cytokines response (higher IL-6, IL-1β and, TNF levels) in both hippocampus and CSF after an instant anti-inflammatory cytokine response in IL-4 over-exposed rats. Moreover, the pro-inflammatory cytokines response appeared earlier in CSF than in hippocampus. The level of each of the pro-inflammatory cytokines in CSF positively correlated with that in hippocampus at the age of postnatal day 42. More microglia numbers/activation and higher M-CSF level in the hippocampus in IL-4 over-exposed rats were also observed. Furthermore, there were more macrophages with inflammatory activation in dural mater of IL-4 over-exposed rats. In sum, neonatal IL-4 over-exposure in rats induces delayed inflammation in CSF, suggesting CSF examination may serve as a potential method in predicting delayed neuroinflammation in brain following neonatal IL-4 over-exposure.
Collapse
Affiliation(s)
- Ling Wang
- Grade 2019, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Haoran Sha
- Grade 2020, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoyi He
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Higher Education Mega Center, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Yinyin Xie
- Grade 2018, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Jiapeng Deng
- Grade 2018, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Jiexuan Chen
- Grade 2020, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, People's Republic of China
| | - Guoying Li
- Guangdong Medical Association, Guangzhou, 510180, Guangdong, China.
| | - Junhua Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Higher Education Mega Center, Guangzhou, 510006, Guangdong, People's Republic of China.
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Higher Education Mega Center, Guangzhou, 510006, Guangdong, People's Republic of China.
| |
Collapse
|
13
|
Devinney MJ, Berger M. Reply to "A Letter Concerning a Role for Blood-Brain Barrier Dysfunction in Delirium following Noncardiac Surgery in Older Adults". Ann Neurol 2024; 95:411-413. [PMID: 37994239 PMCID: PMC11225806 DOI: 10.1002/ana.26834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 11/24/2023]
Affiliation(s)
- Michael J Devinney
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC
| | - Miles Berger
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
14
|
Zhang L, Yao Q, Hu J, Qiu B, Xiao Y, Zhang Q, Zeng Y, Zheng S, Zhang Y, Wan Y, Zheng X, Zeng Q. Hotspots and trends of microglia in Alzheimer's disease: a bibliometric analysis during 2000-2022. Eur J Med Res 2024; 29:75. [PMID: 38268044 PMCID: PMC10807212 DOI: 10.1186/s40001-023-01602-9] [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: 10/29/2023] [Accepted: 12/17/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Alzheimer's disease is one common type of dementia. Numerous studies have suggested a correlation between Alzheimer's disease and inflammation. Microglia mainly participate in the inflammatory response in the brain. Currently, ample evidence has shown that microglia are closely related to the occurrence and development of Alzheimer's disease. OBJECTIVE We opted for bibliometric analysis to comprehensively summarize the advancements in the study of microglia in Alzheimer's disease, aiming to provide researchers with current trends and future research directions. METHODS All articles and reviews pertaining to microglia in Alzheimer's disease from 2000 to 2022 were downloaded through Web of Science Core Collection. The results were subjected to bibliometric analysis using VOSviewer 1.6.18 and CiteSpace 6.1 R2. RESULTS Overall, 7449 publications were included. The number of publications was increasing yearly. The United States has published the most publications. Harvard Medical School has published the most papers of all institutions. Journal of Alzheimer's Disease and Journal of Neuroscience were the journals with the most studies and the most commonly cited, respectively. Mt Heneka is the author with the highest productivity and co-citation. After analysis, the most common keywords are neuroinflammation, amyloid-beta, inflammation, neurodegeneration. Gut microbiota, extracellular vesicle, dysfunction and meta-analysis are the hotspots of research at the present stage and are likely to continue. CONCLUSION NLRP3 inflammasome, TREM2, gut microbiota, mitochondrial dysfunction, exosomes are research hotspots. The relationship between microglia-mediated neuroinflammation and Alzheimer's disease have been the focus of current research and the development trend of future research.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Qiuru Yao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Jinjing Hu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Baizhi Qiu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Yupeng Xiao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Qi Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Yuting Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuqi Zheng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Youao Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yantong Wan
- College of Anesthesiology, Southern Medical University, Guangzhou, China.
| | - Xiaoyan Zheng
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China.
| | - Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
15
|
Jia S, Yang H, Huang F, Fan W. Systemic inflammation, neuroinflammation and perioperative neurocognitive disorders. Inflamm Res 2023; 72:1895-1907. [PMID: 37688642 DOI: 10.1007/s00011-023-01792-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/11/2023] Open
Abstract
Perioperative neurocognitive disorder (PND) is a common disorder following anesthesia and surgery, especially in the elderly. The complex cellular and molecular processes are involved in PND, but the underlying pathogenesis of which remains inconclusive due to conflicting data. A growing body of evidence has been shown that perioperative systemic inflammation plays important roles in the development of PND. We reviewed the relevant literature retrieved by a search in the PubMed database (on July 20, 2023). The search terms used were "delirium", "post operative cognitive dysfunction", "perioperative neurocognitive disorder", "inflammation" and "systemic", alone and in combination. All articles identified were English-language, full-text papers. The ones cited in the review are those that make a substantial contribution to the knowledge about systemic inflammation and PNDs. The aim of this review is to bring together the latest evidence for the understanding of how perioperative systemic inflammation mediates neuroinflammation and brain injury, how the inflammation is regulated and how we can translate these findings into prevention and/or treatment for PND.
Collapse
Affiliation(s)
- Shilin Jia
- Department of Anesthesiology, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Hui Yang
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Fang Huang
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Wenguo Fan
- Department of Anesthesiology, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| |
Collapse
|
16
|
de la Monte SM, Tong M, Hapel AJ. Concordant and Discordant Cerebrospinal Fluid and Plasma Cytokine and Chemokine Responses in Mild Cognitive Impairment and Early-Stage Alzheimer's Disease. Biomedicines 2023; 11:2394. [PMID: 37760836 PMCID: PMC10525668 DOI: 10.3390/biomedicines11092394] [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: 07/21/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Neuroinflammation may be a pathogenic mediator and biomarker of neurodegeneration at the boundary between mild cognitive impairment (MCI) and early-stage Alzheimer's disease (AD). Whether neuroinflammatory processes are endogenous to the central nervous system (CNS) or originate from systemic (peripheral blood) sources could impact strategies for therapeutic intervention. To address this issue, we measured cytokine and chemokine immunoreactivities in simultaneously obtained lumbar puncture cerebrospinal fluid (CSF) and serum samples from 39 patients including 18 with MCI or early AD and 21 normal controls using a 27-plex XMAP bead-based enzyme-linked immunosorbent assay (ELISA). The MCI/AD combined group had significant (p < 0.05 or better) or statistically trend-wise (0.05 ≤ p ≤ 0.10) concordant increases in CSF and serum IL-4, IL-5, IL-9, IL-13, and TNF-α and reductions in GM-CSF, b-FGF, IL-6, IP-10, and MCP-1; CSF-only increases in IFN-y and IL-7 and reductions in VEGF and IL-12p70; serum-only increases in IL-1β, MIP-1α, and eotaxin and reductions in G-CSF, IL-2, IL-8 and IL-15; and discordant CSF-serum responses with reduced CSF and increased serum PDGF-bb, IL-17a, and RANTES. The results demonstrate simultaneously parallel mixed but modestly greater pro-inflammatory compared to anti-inflammatory or neuroprotective responses in CSF and serum. In addition, the findings show evidence that several cytokines and chemokines are selectively altered in MCI/AD CSF, likely corresponding to distinct neuroinflammatory responses unrelated to systemic pathologies. The aggregate results suggest that early management of MCI/AD neuroinflammation should include both anti-inflammatory and pro-neuroprotective strategies to help prevent disease progression.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology (Neuropathology), Neurology, and Neurosurgery, Rhode Island Hospital, The Alpert Medical School of Brown University, Providence, RI 02903, USA
- Department of Medicine, Rhode Island Hospital, The Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, The Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Andrew J. Hapel
- Department of Genome Biology, John Curtin School of Medical Research, Australian National University, Canberra 2601, Australia;
| |
Collapse
|
17
|
Ma Y, Farny NG. Connecting the dots: Neuronal senescence, stress granules, and neurodegeneration. Gene 2023; 871:147437. [PMID: 37084987 PMCID: PMC10205695 DOI: 10.1016/j.gene.2023.147437] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/09/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
Cellular senescence increases with aging. While senescence is associated with an exit of the cell cycle, there is ample evidence that post-mitotic cells including neurons can undergo senescence as the brain ages, and that senescence likely contributes significantly to the progression of neurodegenerative diseases (ND) such as Alzheimer's Disease (AD) and Amyotrophic Lateral Sclerosis (ALS). Stress granules (SGs) are stress-induced cytoplasmic biomolecular condensates of RNA and proteins, which have been linked to the development of AD and ALS. The SG seeding hypothesis of NDs proposes that chronic stress in aging neurons results in static SGs that progress into pathological aggregates Alterations in SG dynamics have also been linked to senescence, though studies that link SGs and senescence in the context of NDs and the aging brain have not yet been performed. In this Review, we summarize the literature on senescence, and explore the contribution of senescence to the aging brain. We describe senescence phenotypes in aging neurons and glia, and their links to neuroinflammation and the development of AD and ALS. We further examine the relationships of SGs to senescence and to ND. We propose a new hypothesis that neuronal senescence may contribute to the mechanism of SG seeding in ND by altering SG dynamics in aged cells, thereby providing additional aggregation opportunities within aged neurons.
Collapse
Affiliation(s)
- Yizhe Ma
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Natalie G Farny
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, USA.
| |
Collapse
|
18
|
Nakagawa Y, Yamada S. The Relationships Among Metal Homeostasis, Mitochondria, and Locus Coeruleus in Psychiatric and Neurodegenerative Disorders: Potential Pathogenetic Mechanism and Therapeutic Implications. Cell Mol Neurobiol 2023; 43:963-989. [PMID: 35635600 PMCID: PMC11414457 DOI: 10.1007/s10571-022-01234-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 05/15/2022] [Indexed: 11/03/2022]
Abstract
While alterations in the locus coeruleus-noradrenergic system are present during early stages of neuropsychiatric disorders, it is unclear what causes these changes and how they contribute to other pathologies in these conditions. Data suggest that the onset of major depressive disorder and schizophrenia is associated with metal dyshomeostasis that causes glial cell mitochondrial dysfunction and hyperactivation in the locus coeruleus. The effect of the overactive locus coeruleus on the hippocampus, amygdala, thalamus, and prefrontal cortex can be responsible for some of the psychiatric symptoms. Although locus coeruleus overactivation may diminish over time, neuroinflammation-induced alterations are presumably ongoing due to continued metal dyshomeostasis and mitochondrial dysfunction. In early Alzheimer's and Parkinson's diseases, metal dyshomeostasis and mitochondrial dysfunction likely induce locus coeruleus hyperactivation, pathological tau or α-synuclein formation, and neurodegeneration, while reduction of glymphatic and cerebrospinal fluid flow might be responsible for β-amyloid aggregation in the olfactory regions before the onset of dementia. It is possible that the overactive noradrenergic system stimulates the apoptosis signaling pathway and pathogenic protein formation, leading to further pathological changes which can occur in the presence or absence of locus coeruleus hypoactivation. Data are presented in this review indicating that although locus coeruleus hyperactivation is involved in pathological changes at prodromal and early stages of these neuropsychiatric disorders, metal dyshomeostasis and mitochondrial dysfunction are critical factors in maintaining ongoing neuropathology throughout the course of these conditions. The proposed mechanistic model includes multiple pharmacological sites that may be targeted for the treatment of neuropsychiatric disorders commonly.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| |
Collapse
|
19
|
Zarate SM, Huntington TE, Bagher P, Srinivasan R. Aging reduces calreticulin expression and alters spontaneous calcium signals in astrocytic endfeet of the mouse dorsolateral striatum. NPJ AGING 2023; 9:5. [PMID: 37002232 PMCID: PMC10066375 DOI: 10.1038/s41514-023-00102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 03/15/2023] [Indexed: 04/03/2023]
Abstract
Aging-related impairment of the blood brain barrier (BBB) and neurovascular unit (NVU) increases the risk for neurodegeneration. Among various cells that participate in BBB and NVU function, calcium signals in astrocytic endfeet are crucial for maintaining BBB and NVU integrity. To assess if aging is associated with altered calcium signals within astrocytic endfeet of the dorsolateral striatum (DLS), we expressed GCaMP6f in DLS astrocytes of young (3-4 months), middle-aged (12-15 months) and aging (20-30 months) mice. Compared to endfeet in young mice, DLS endfeet in aging mice demonstrated decreased calreticulin expression, and alterations to both spontaneous membrane-associated and mitochondrial calcium signals. While young mice required both extracellular and endoplasmic reticulum calcium sources for endfoot signals, middle-aged and aging mice showed heavy dependence on endoplasmic reticulum calcium. Thus, astrocytic endfeet show significant changes in calcium buffering and sources throughout the lifespan, which is important for understanding mechanisms by which aging impairs the BBB and NVU.
Collapse
Affiliation(s)
- Sara M Zarate
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
| | - Taylor E Huntington
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, 8447 Riverside Pkwy, Bryan, TX, 77807, USA
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, 77843, USA
| | - Pooneh Bagher
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, 8447 Riverside Pkwy, Bryan, TX, 77807, USA.
- Texas A&M Institute for Neuroscience (TAMIN), Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
20
|
Monteiro AR, Barbosa DJ, Remião F, Silva R. Alzheimer’s disease: insights and new prospects in disease pathophysiology, biomarkers and disease-modifying drugs. Biochem Pharmacol 2023; 211:115522. [PMID: 36996971 DOI: 10.1016/j.bcp.2023.115522] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases that affect millions of people worldwide, with both prevalence and incidence increasing with age. It is characterized by cognitive decline associated, specifically, with degeneration of cholinergic neurons. The problem of this disease is even more fundamental as the available therapies remain fairly limited and mainly focused on symptoms' relief. Although the aetiology of the disease remains elusive, two main pathological hallmarks are described: i) presence of neurofibrillary tangles formed by unfolded protein aggregates (hyperphosphorylated Tau protein) and ii) presence of extracellular aggregates of amyloid-beta peptide. Given the complexity surrounding the pathogenesis of the disease, several potential targets have been highlighted and interrelated upon its progression, such as oxidative stress and the accumulation of metal ions. Thus, advances have been made on the development of innovative multitarget therapeutical compounds to delay the disease progression and restore cell function. This review focuses the ongoing research on new insights and emerging disease-modifying drugs for AD treatment. Furthermore, classical and novel potential biomarkers for early diagnosis of the disease, and their role in assisting on the improvement of targeted therapies will also be approached.
Collapse
Affiliation(s)
- Ana R Monteiro
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniel J Barbosa
- TOXRUN - Toxicology Research Unit, Department of Sciences, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Fernando Remião
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, Porto University, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
21
|
Martins GL, Ferreira CN, Palotás A, Rocha NP, Reis HJ. Role of Oxysterols in the Activation of the NLRP3 Inflammasome as a Potential Pharmacological Approach in Alzheimer's Disease. Curr Neuropharmacol 2023; 21:202-212. [PMID: 35339182 PMCID: PMC10190144 DOI: 10.2174/1570159x20666220327215245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/04/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, is a complex clinical condition with multifactorial origin posing a major burden to health care systems across the world. Even though the pathophysiological mechanisms underlying the disease are still unclear, both central and peripheral inflammation has been implicated in the process. Piling evidence shows that the nucleotide-binding domain, leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome is activated in AD. As dyslipidemia is a risk factor for dementia, and cholesterol can also activate the inflammasome, a possible link between lipid levels and the NLRP3 inflammasome has been proposed in Alzheimer's. It is also speculated that not only cholesterol but also its metabolites, the oxysterols, may be involved in AD pathology. In this context, mounting data suggest that NLRP3 inflammasome activity can be modulated by different peripheral nuclear receptors, including liver-X receptors, which present oxysterols as endogenous ligands. In light of this, the current review explores whether the activation of NLRP3 by nuclear receptors, mediated by oxysterols, may also be involved in AD and could serve as a potential pharmacological avenue in dementia.
Collapse
Affiliation(s)
- Gabriela L. Martins
- Laboratório Neurofarmacologia, Departamento de Farmacologia, ICB-UFMG, Belo Horizonte MG, 31270 - 901, Brazil
| | | | - András Palotás
- Kazan Federal University, Kazan, Russia
- Asklepios Med, Szeged, Hungary
| | - Natália P. Rocha
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Helton J. Reis
- Laboratório Neurofarmacologia, Departamento de Farmacologia, ICB-UFMG, Belo Horizonte MG, 31270 - 901, Brazil
| |
Collapse
|
22
|
Kann O, Almouhanna F, Chausse B. Interferon γ: a master cytokine in microglia-mediated neural network dysfunction and neurodegeneration. Trends Neurosci 2022; 45:913-927. [PMID: 36283867 DOI: 10.1016/j.tins.2022.10.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Traditionally, lymphocytic interferon γ (IFN-γ) was considered to be a simple 'booster' of proinflammatory responses by microglia (brain-resident macrophages) during bacterial or viral infection. Recent slice culture (in situ) and in vivo studies suggest, however, that IFN-γ has a unique role in microglial activation. Priming by IFN-γ results in proliferation (microgliosis), enhanced synapse elimination, and moderate nitric oxide release sufficient to impair synaptic transmission, gamma rhythm activity, and cognitive functions. Moreover, IFN-γ is pivotal for driving Toll-like receptor (TLR)-activated microglia into neurotoxic phenotypes that induce energetic and oxidative stress, severe network dysfunction, and neuronal death. Pharmacological targeting of activated microglia could be beneficial during elevated IFN-γ levels, blood-brain barrier leakage, and parenchymal T lymphocyte infiltration associated with, for instance, encephalitis, multiple sclerosis, and Alzheimer's disease.
Collapse
Affiliation(s)
- Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, D-69120 Heidelberg, Germany.
| | - Fadi Almouhanna
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Bruno Chausse
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
23
|
Differential compartmentalization of myeloid cell phenotypes and responses towards the CNS in Alzheimer's disease. Nat Commun 2022; 13:7210. [PMID: 36418303 PMCID: PMC9684147 DOI: 10.1038/s41467-022-34719-2] [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: 02/25/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022] Open
Abstract
Myeloid cells are suggested as an important player in Alzheimer´s disease (AD). However, its continuum of phenotypic and functional changes across different body compartments and their use as a biomarker in AD remains elusive. Here, we perform multiple state-of-the-art analyses to phenotypically and metabolically characterize immune cells between peripheral blood (n = 117), cerebrospinal fluid (CSF, n = 117), choroid plexus (CP, n = 13) and brain parenchyma (n = 13). We find that CSF cells increase expression of markers involved in inflammation, phagocytosis, and metabolism. Changes in phenotype of myeloid cells from AD patients are more pronounced in CP and brain parenchyma and upon in vitro stimulation, suggesting that AD-myeloid cells are more vulnerable to environmental changes. Our findings underscore the importance of myeloid cells in AD and the detailed characterization across body compartments may serve as a resource for future studies focusing on the assessment of these cells as biomarkers in AD.
Collapse
|
24
|
Zheng S, Pan L, Hou J, Liao A, Hou Y, Yu G, Li X, Yuan Y, Dong Y, Zhao P, Zhang J, Hu Z, Hui M, Cao J, Huang JH. The role of wheat embryo globulin nutrients in improving cognitive dysfunction in AD rats. Food Funct 2022; 13:9856-9867. [PMID: 36047913 DOI: 10.1039/d2fo00815g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neuroinflammation and intestinal microbiota cause pathological progression of Alzheimer's disease (AD), leading to neurodegeneration and cognitive decline. This study investigates the effects of wheat embryo globulin nutrient (WEGN) on depression, neuroinflammation, and intestinal microbial disorder caused by AD and its protective mechanism on cognitive impairment. Results demonstrated that rats in the WEGN group have lower feed intake but higher body weight than those in the control group. Notably, rats in the WEGN group have a higher number of cross grids and uprights and a smaller amount of fecal particles than those in the control group. Biochemical examinations revealed that rats in the WEGN group had lower expression of interleukin-1β, interleukin-6, and tumor necrosis factor α in hippocampus tissue and the expression of genes and proteins related to the TLR4/MyD88/NF-κB signaling pathway in AD rats was down-regulated compared to those in the control group. The 16S rRNA gene sequencing results demonstrated that WEGN treatment inhibits the increase of Erysipelotrichaceae, Erysipelatoclostridium, Erysipelotrichaceae, Corynebacterium, and Frisingicoccus, and the reduction of Lactobacillus in AD rats. WEGN has potential value as a practical food in alleviating neuroinflammation-related diseases such as AD.
Collapse
Affiliation(s)
- Shuainan Zheng
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Long Pan
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Jianguang Hou
- Workstation of Zhongyuan Scholars of Henan Province, Henan Yangshao Liquor Co., Ltd., Mianchi Xian, 472400, PR China
| | - Aimei Liao
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Yinchen Hou
- College of Food and Biological Engineering, Henan University of Animal Husbandry and Economy, Zhengzhou 450044, PR China
| | - Guanghai Yu
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Xiaoxiao Li
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Yongjian Yuan
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Yuqi Dong
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Penghui Zhao
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Jie Zhang
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Zheyuan Hu
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Ming Hui
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Jian Cao
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China.
| | - Ji-Hong Huang
- Henan Provincial Key Laboratory of Biological Processing and Nutritional Function of Wheat, School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, PR China. .,School of Food and Pharmacy, Xuchang University, Xuchang 461000, PR China.,State Key Laboratory of Crop Stress Adaptation and Improvement, College of Agriculture, Henan University, Kaifeng 475004, China.
| |
Collapse
|
25
|
Sun Q, Ni J, Wei M, Long S, Li T, Fan D, Lu T, Shi J, Tian J. Plasma β-amyloid, tau, neurodegeneration biomarkers and inflammatory factors of probable Alzheimer’s disease dementia in Chinese individuals. Front Aging Neurosci 2022; 14:963845. [PMID: 36062146 PMCID: PMC9433929 DOI: 10.3389/fnagi.2022.963845] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundPlasma-derived β-amyloid, tau, and neurodegeneration (ATN) biomarkers can accurately diagnose Alzheimer’s disease (AD) and predict its progression. Few studies have investigated the relationship between plasma biomarkers and changes in plasma inflammatory markers in clinically diagnosed AD.MethodsSeventy-four participants were recruited, including 30 mild-to-moderate AD dementia patients and 44 normal controls (NC). All participants underwent neuropsychological testing and blood sampling for biomarker testing. AD was clinically diagnosed according to the National Institute on Aging-Alzheimer’s Association (NIA-AA) core criteria and required age-mismatched hippocampal atrophy. We performed Single Molecule Array (Simoa), an ultra-sensitive enzyme-linked immunosorbent assay (ELISA), to examine plasma ATN markers, including β-amyloid (Aβ) 40, Aβ42, p-tau181, total (t)-tau, neurofilament protein light chain (NfL), and inflammatory factors (TNF-α, IL-1β, IL-6, and IL-8).ResultsThe level of the plasma Aβ42/Aβ40 ratio was significantly declined and the levels of the plasma p-tau181, NfL and TNF-α were significantly higher in the AD group than the NC group, but there was no significant difference in the levels of plasma t-tau, IL-1β, IL-6, and IL-8 between the AD and NC groups. The levels of plasma p-tau181, NfL, Aβ42/Aβ40 ratio, and TNF-α were all associated with impairments in multiple cognitive domains. Among them, the plasma Aβ42/Aβ40 ratio, and the p-tau181 and TNF-α levels were associated with impairments in global cognition, memory, and visuospatial abilities, but not with executive function, only plasma NfL level was associated with executive function. Plasma NfL showed higher diagnostic performance in AD than in NC individuals (AUC = 0.833). A combined diagnostic prediction model of plasma Aβ42/Aβ40 ratio, p-tau 181, and NfL had the highest value than each factor alone (AUC = 0.902),with a sensitivity and specificity of 0.867 and 0.886, respectively.ConclusionThe levels of plasma ATN biomarkers (Aβ42/Aβ40 ratio, p-tua181, and NfL) were significantly changed in clinically diagnosed AD patients and they all associated with different domains of cognitive impairment. Plasma ATN biomarkers better differentiate mild-to-moderate AD dementia from NC when they are incorporated into diagnostic models together rather than individually. Plasma ATN biomarkers have the potential to be a screening tool for AD. However, the expression of inflammatory factors in AD patients requires further research.
Collapse
Affiliation(s)
- Qingling Sun
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingnian Ni
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingqing Wei
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Siwei Long
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Li
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Tao Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Shi
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Jing Shi,
| | - Jinzhou Tian
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Jinzhou Tian,
| |
Collapse
|
26
|
Koca S, Kiris I, Sahin S, Cinar N, Karsidag S, Hanagasi HA, Yildiz GB, Tarik Baykal A. Decreased levels of cytokines implicate altered immune response in plasma of moderate-stage Alzheimer's disease patients. Neurosci Lett 2022; 786:136799. [PMID: 35842208 DOI: 10.1016/j.neulet.2022.136799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/18/2022]
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disease characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. However, increasing evidence suggests that the pathogenesis of the disease is associated with peripheral inflammation. Here, we aimed to determine plasma concentrations of multiple cytokines and chemokines from moderate-stage AD and age-matched controls. Changes in a total of 20 cytokines and chemokines in plasma of moderate-stage AD were evaluated by using quantitative microarray. Six of them, namely MCP-1, MIP-1a, MIP-1b, MMP-9, RANTES, and VEGF, were found to be significantly reduced in moderate-stage AD patients (n = 25) in comparison to age-matched and non-demented controls (n = 25). However, GM-CSF, GRO-α/β/γ, IFN- γ, IL-1α, IL-1β, IL-10, IL-12 p70, IL-13, IL-2, IL- 4, IL-5, IL-6, IL-8, and TNF-α showed no significant differences between the patient and control groups. On the contrary to previous early-stage AD studies that show increased plasma cytokine/chemokine levels, our results indicate that inflammatory plasma molecules are reduced in moderate-stage AD. This finding points out the reduced immune responsiveness, which is known to be directly correlated to the degree of AD.
Collapse
Affiliation(s)
- Sebile Koca
- Department of Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Irem Kiris
- Department of Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Sevki Sahin
- Department of Neurology, Faculty of Medicine, Maltepe University, Istanbul, Turkey
| | - Nilgun Cinar
- Department of Neurology, Faculty of Medicine, Maltepe University, Istanbul, Turkey
| | - Sibel Karsidag
- Department of Neurology, Faculty of Medicine, Maltepe University, Istanbul, Turkey
| | - Hasmet A Hanagasi
- Department of Neurology, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Gulsen B Yildiz
- Department of Neurology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Ahmet Tarik Baykal
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
| |
Collapse
|
27
|
The "Cerebrospinal Fluid Sink Therapeutic Strategy" in Alzheimer's Disease-From Theory to Design of Applied Systems. Biomedicines 2022; 10:biomedicines10071509. [PMID: 35884814 PMCID: PMC9313192 DOI: 10.3390/biomedicines10071509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is a global health problem, with incidence and prevalence considered to increase during the next decades. However, no currently available effective treatment exists despite numerous clinical trials in progress. Moreover, although many hypotheses are accepted regarding the pathophysiological mechanisms of AD onset and evolution, there are still many unknowns about the disorder. A relatively new approach, based on the amyloid-beta dynamics among different biological compartments, is currently intensely discussed, as it seems to offer a promising solution with significant therapeutic impact. Known as the “cerebrospinal-fluid-sink therapeutic strategy”, part of the “three-sink therapeutic strategy”, this theoretical model focuses on the dynamics of amyloid-beta among the three main liquid compartments of the human body, namely blood, cerebrospinal fluid, and the (brain) interstitial fluid. In this context, this article aims to describe in detail the abovementioned hypothesis, by reviewing in the first part the most relevant anatomical and physiological aspects of amyloid-beta dynamics. Subsequently, explored therapeutic strategies based on the clearance of amyloid-beta from the cerebrospinal fluid level are presented, additionally highlighting their limitations. Finally, the originality and novelty of this work rely on the research experience of the authors, who focus on implantable devices and their utility in AD treatment.
Collapse
|
28
|
Saleh MAA, Bloemberg JS, Elassaiss-Schaap J, de Lange ECM. Drug Distribution in Brain and Cerebrospinal Fluids in Relation to IC 50 Values in Aging and Alzheimer's Disease, Using the Physiologically Based LeiCNS-PK3.0 Model. Pharm Res 2022; 39:1303-1319. [PMID: 35606598 PMCID: PMC9246802 DOI: 10.1007/s11095-022-03281-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/27/2022] [Indexed: 12/02/2022]
Abstract
Background Very little knowledge exists on the impact of Alzheimer’s disease on the CNS target site pharmacokinetics (PK). Aim To predict the CNS PK of cognitively healthy young and elderly and of Alzheimer’s patients using the physiologically based LeiCNS-PK3.0 model. Methods LeiCNS-PK3.0 was used to predict the PK profiles in brain extracellular (brainECF) and intracellular (brainICF) fluids and cerebrospinal fluid of the subarachnoid space (CSFSAS) of donepezil, galantamine, memantine, rivastigmine, and semagacestat in young, elderly, and Alzheimer’s patients. The physiological parameters of LeiCNS-PK3.0 were adapted for aging and Alzheimer’s based on an extensive literature search. The CNS PK profiles at plateau for clinical dose regimens were related to in vitro IC50 values of acetylcholinesterase, butyrylcholinesterase, N-methyl-D-aspartate, or gamma-secretase. Results The PK profiles of all drugs differed between the CNS compartments regarding plateau levels and fluctuation. BrainECF, brainICF and CSFSAS PK profile relationships were different between the drugs. Aging and Alzheimer’s had little to no impact on CNS PK. Rivastigmine acetylcholinesterase IC50 values were not reached. Semagacestat brain PK plateau levels were below the IC50 of gamma-secretase for half of the interdose interval, unlike CSFSAS PK profiles that were consistently above IC50. Conclusion This study provides insights into the relations between CNS compartments PK profiles, including target sites. CSFSAS PK appears to be an unreliable predictor of brain PK. Also, despite extensive changes in blood-brain barrier and brain properties in Alzheimer’s, this study shows that the impact of aging and Alzheimer’s pathology on CNS distribution of the five drugs is insignificant. Supplementary Information The online version contains supplementary material available at 10.1007/s11095-022-03281-3.
Collapse
Affiliation(s)
- Mohammed A A Saleh
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Julia S Bloemberg
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Jeroen Elassaiss-Schaap
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
- PD-value B.V., Houten, The Netherlands
| | - Elizabeth C M de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
29
|
Sternberg Z, Podolsky R, Nir A, Yu J, Nir R, Halvorsen SW, Quinn JF, Kaye J, Kolb C. Elevated spermidine serum levels in mild cognitive impairment, a potential biomarker of progression to Alzheimer dementia, a pilot study. J Clin Neurosci 2022; 100:169-174. [PMID: 35487023 DOI: 10.1016/j.jocn.2022.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/07/2022] [Accepted: 04/22/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND/AIMS There is a close link between iron and polyamine biosynthesis and metabolism. In a recent study, we reported alterations in the serum levels of hepcidin and other iron-related proteins in Alzheimer's disease (AD) patients (Sternberg et al., 2017). Based on these findings, this pilot study compared serum levels of one of the polyamines, Spermidine, between AD, mild cognitive impairment (MCI), and control subjects, correlating the levels with the existing clinical and neuroimaging data. METHODS This cross-sectional study measured Spermidine levels in frozen serum samples of 43 AD patients, 12 MCI patients, and 21 age-matched controls, provided by the Oregon Alzheimer's Disease Center Bio-repository, using enzyme-linked immunosorbent assay. RESULTS MCI patients showed significantly higher mean Spermidine serum levels compared to controls (P = 0.01), with a non-significant trend for higher Spermidine serum levels in pure AD (P = 0.08) participants compared to controls. Spermidine serum levels correlated with the values of cognitive assessment tests including MMSE (r = -0.705, P = 0.003), CDR (r = 0.751, P = 0.002), and CDR-SOB (r = 0.704, P = 0.007), in "pure" AD subgroup, suggesting that higher Spermidine serum levels in MCI can be a potential biomarker of conversion to dementia in subjects with AD underlying pathology. Furthermore, Spermidine serum levels correlated with serum levels of the chief iron regulatory protein, hepcidin in AD participants with a more advanced disease stage, indicated by MMSE (strata of 8-19, P = 0.02), and CDR-SOB (strata of 6-12, P = 0.03). CONCLUSION Studies with larger cohort are warranted for defining the role of Spermidine in AD pathophysiology, and the utility of polyamines as biomarkers of progression of MCI to AD.
Collapse
Affiliation(s)
- Zohara Sternberg
- Department of Neurology, Stroke Center, Buffalo Medical Center, Buffalo, NY, USA.
| | - Rebecca Podolsky
- Department of Neurology, Stroke Center, Buffalo Medical Center, Buffalo, NY, USA
| | | | - Jihnhee Yu
- Department of Biostatistics, University of Buffalo, Buffalo, NY, USA
| | | | - Stanley W Halvorsen
- Department of Pharmacology and Toxicology, University of Buffalo, Buffalo, NY, USA
| | - Joseph F Quinn
- Layton Aging & Alzheimer's Research Center, Oregon Health and Science University, Portland, OR, USA
| | - Jeffrey Kaye
- Layton Aging & Alzheimer's Research Center, Oregon Health and Science University, Portland, OR, USA
| | - Channa Kolb
- Department of Neurology, Stroke Center, Buffalo Medical Center, Buffalo, NY, USA
| |
Collapse
|
30
|
Aksnes M, Aass HCD, Tiiman A, Terenius L, Bogdanović N, Vukojević V, Knapskog AB. Serum Amyloidogenic Nanoplaques and Cytokines in Alzheimer's Disease: Pilot Study in a Small Naturalistic Memory Clinic Cohort. J Alzheimers Dis 2022; 86:1459-1470. [PMID: 35213378 PMCID: PMC9108575 DOI: 10.3233/jad-215504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Neuroinflammation is a central component of Alzheimer’s disease (AD) and correlates closely with amyloid pathology. Markers of inflammation such as cytokines, and amyloidogenic aggregates, so-called nanoplaques, are both promising biomarker candidates for AD. We have previously shown that there is a relationship between the levels of nanoplaques and cytokines in cerebrospinal fluid, but it is unknown whether this association extends to serum. Objective: Investigate in a naturalistic memory clinic cohort whether the associations between nanoplaques and cytokines in the cerebrospinal fluid extends to serum. Methods: We collected serum from 49 patients assessed for cognitive complaints at the Oslo University Hospital Memory Clinic (15 with clinical AD). We assessed the levels of serum nanoplaques with the novel Thioflavin-T fluorescence correlation spectroscopy (ThT-FCS) assay. Serum levels of nine cytokines (eotaxin-1, granulocyte colony-stimulating factor [G-CSF], interleukin [IL]-6, IL-7, IL-8, monocyte chemoattractant protein-1 (MCP-1), gamma induced protein 10 (IP-10), macrophage inflammatory protein [MIP]-1α, and MIP-1β) were quantified with a multiplex assay and read on a Luminex IS 200 instrument. Results: Serum nanoplaques were not increased in clinical AD patients compared to non-AD memory clinic patients and nanoplaques were not associated with any cytokines. The cytokines IL-8 and G-CSF were increased in patients with clinical AD compared to non-AD patients. Conclusion: In this small pilot study, serum nanoplaques were not associated with serum cytokines. Nanoplaque levels could not be used to separate clinical AD patients from non-AD patients in this unselected memory clinic cohort.
Collapse
Affiliation(s)
- Mari Aksnes
- Department of Geriatric Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | | | - Ann Tiiman
- Department of Clinical Neurosciences (CNS), Center for Molecular Medicine CMM L8:01, Karolinska Institutet, Stockholm, Sweden
| | - Lars Terenius
- Department of Clinical Neurosciences (CNS), Center for Molecular Medicine CMM L8:01, Karolinska Institutet, Stockholm, Sweden
| | - Nenad Bogdanović
- Department of Geriatric Medicine, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway.,Department of Neurobiology, Care Science and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden
| | - Vladana Vukojević
- Department of Clinical Neurosciences (CNS), Center for Molecular Medicine CMM L8:01, Karolinska Institutet, Stockholm, Sweden
| | - Anne-Brita Knapskog
- Department of Geriatric Medicine, The Memory Clinic, Oslo University Hospital, Norway
| |
Collapse
|
31
|
Xiong W, Zhao X, Xu Q, Wei G, Zhang L, Fan Y, Wen L, Liu Y, Zhang T, Zhang L, Tong Y, Yin Q, Zhang TE, Yan Z. Qisheng Wan formula ameliorates cognitive impairment of Alzheimer's disease rat via inflammation inhibition and intestinal microbiota regulation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114598. [PMID: 34492320 DOI: 10.1016/j.jep.2021.114598] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Qisheng Wan formula (QWF) was first described in the book Sheng Ji Zong Lu in 1117. The book states that QWF can cure forgetfulness, improve the mind, and make people smart. Hence, QWF has been widely used to treat patients with forgetfulness or dementia. QWF, a classic Chinese formulation, comprises seven herbal drugs: the sclerotium of Poria cocos (Schw.) Wolf, bark of Cinnamomum cassia Presl, root of Polygala tenuifolia Willd., root and rhizome of Panax ginseng C. A. Mey., root of Asparagus cochinchinensis (Lour.) Merr., root and rhizome of Acorus tatarinowii Schott, and root bark of Lycium chinense Mill. AIM OF THE STUDY This study aimed to utilize modern pharmacological methods to evaluate the therapeutic effects and explore the underlying mechanism of QWF action on rats with Alzheimer's disease (AD). MATERIALS AND METHODS The chemical profile of QWF was characterized using ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. The AD rat model was established via a bilateral intraventricular injection of amyloid-β (1-42) (Aβ1-42). The rats were subsequently treated daily with QWF for 4 weeks. The Morris water maze test was performed to evaluate the cognition processes in the rats, whereas histological changes in the hippocampus were observed using hematoxylin and eosin staining. The expression levels of Aβ1-42, nuclear factor-kappa B (NF-κB), tumor necrosis factor (TNF)-α, and interleukin (IL)-6 in the hippocampus and colon were assessed. Moreover, the diversity and composition of the intestinal microbiota were analyzed using 16S rDNA gene sequencing. RESULTS One hundred and fourteen compounds were characterized in QWF. QWF significantly ameliorated the cognition processes and histopathological damages due to AD in rats by decreasing the deposition of Aβ1-42 and downregulating the expression of NF-κB, TNF-α, and IL-6. QWF also modulated changes in the diversity and composition of intestinal microbiota to suppress the relative abundance of inflammation-associated microbiota. CONCLUSION This study showed that QWF can suppress proinflammatory factors and modulate the intestinal microbiota in AD rats.
Collapse
Affiliation(s)
- Wei Xiong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Xiaoqin Zhao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Qing Xu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Guihua Wei
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Liudai Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Yuqing Fan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Lingmiao Wen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Yanjun Liu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Tinglan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Li Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Yan Tong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| | - Qiaozhi Yin
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| | - Tian-E Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China.
| | - Zhiyong Yan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, PR China.
| |
Collapse
|
32
|
Salmina AB, Malinovskaya NA, Morgun AV, Khilazheva ED, Uspenskaya YA, Illarioshkin SN. Reproducibility of developmental neuroplasticity in in vitro brain tissue models. Rev Neurosci 2022; 33:531-554. [PMID: 34983132 DOI: 10.1515/revneuro-2021-0137] [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: 10/13/2021] [Accepted: 12/13/2021] [Indexed: 11/15/2022]
Abstract
The current prevalence of neurodevelopmental, neurodegenerative diseases, stroke and brain injury stimulates studies aimed to identify new molecular targets, to select the drug candidates, to complete the whole set of preclinical and clinical trials, and to implement new drugs into routine neurological practice. Establishment of protocols based on microfluidics, blood-brain barrier- or neurovascular unit-on-chip, and microphysiological systems allowed improving the barrier characteristics and analyzing the regulation of local microcirculation, angiogenesis, and neurogenesis. Reconstruction of key mechanisms of brain development and even some aspects of experience-driven brain plasticity would be helpful in the establishment of brain in vitro models with the highest degree of reliability. Activity, metabolic status and expression pattern of cells within the models can be effectively assessed with the protocols of system biology, cell imaging, and functional cell analysis. The next generation of in vitro models should demonstrate high scalability, 3D or 4D complexity, possibility to be combined with other tissues or cell types within the microphysiological systems, compatibility with bio-inks or extracellular matrix-like materials, achievement of adequate vascularization, patient-specific characteristics, and opportunity to provide high-content screening. In this review, we will focus on currently available and prospective brain tissue in vitro models suitable for experimental and preclinical studies with the special focus on models enabling 4D reconstruction of brain tissue for the assessment of brain development, brain plasticity, and drug kinetics.
Collapse
Affiliation(s)
- Alla B Salmina
- Laboratory of Experimental Brain Cytology, Research Center of Neurology, Volokolamskoe Highway 80, Moscow, 125367, Russia.,Research Institute of Molecular Medicine & Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, P. Zhelenzyaka str., 1, Krasnoyarsk 660022, Russia
| | - Natalia A Malinovskaya
- Research Institute of Molecular Medicine & Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, P. Zhelenzyaka str., 1, Krasnoyarsk 660022, Russia
| | - Andrey V Morgun
- Department of Ambulatory Pediatrics, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, P. Zheleznyaka str., 1, Krasnoyarsk 660022, Russia
| | - Elena D Khilazheva
- Research Institute of Molecular Medicine & Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, P. Zhelenzyaka str., 1, Krasnoyarsk 660022, Russia
| | - Yulia A Uspenskaya
- Research Institute of Molecular Medicine & Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, P. Zhelenzyaka str., 1, Krasnoyarsk 660022, Russia
| | - Sergey N Illarioshkin
- Department of Brain Studies, Research Center of Neurology, Volokolamskoe Highway, 80, Moscow 125367, Russia
| |
Collapse
|
33
|
Chouhan JK, Püntener U, Booth SG, Teeling JL. Systemic Inflammation Accelerates Changes in Microglial and Synaptic Markers in an Experimental Model of Chronic Neurodegeneration. Front Neurosci 2022; 15:760721. [PMID: 35058740 PMCID: PMC8764443 DOI: 10.3389/fnins.2021.760721] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/30/2021] [Indexed: 12/01/2022] Open
Abstract
Bacterial infections are a common cause of morbidity and mortality in the elderly, and particularly in individuals with a neurodegenerative disease. Experimental models of neurodegeneration have shown that LPS-induced systemic inflammation increases neuronal damage, a process thought to be mediated by activation of "primed" microglia. The effects of a real systemic bacterial infection on the innate immune cells in the brain and neuronal networks are less well described, and therefore, in this study we use the ME7 prion model to investigate the alterations in microglia activation and phenotype and synaptic markers in response to a low grade, live bacterial infection. Mice with or without a pre-existing ME7 prion-induced neurodegenerative disease were given a single systemic injection of live Salmonella typhimurium at early or mid-stage of disease progression. Immune activation markers CD11b and MHCII and pro-inflammatory cytokines were analyzed 4 weeks post-infection. Systemic infection with S. typhimurium resulted in an exaggerated inflammatory response when compared to ME7 prion mice treated with saline. These changes to inflammatory markers were most pronounced at mid-stage disease. Analysis of synaptic markers in ME7 prion mice revealed a significant reduction of genes that are associated with early response in synaptic plasticity, extracellular matrix structure and post-synaptic density, but no further reduction following systemic infection. In contrast, analysis of activity-related neuronal receptors involved in development of learning and memory, such as Grm1 and Grin2a, showed a significant decrease in response to systemic bacterial challenge. These changes were observed early in the disease progression and associated with reduced burrowing activity. The exaggerated innate immune activation and altered expression of genes linked to synaptic plasticity may contribute to the onset and/or progression of neurodegeneration.
Collapse
Affiliation(s)
| | | | | | - Jessica L. Teeling
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
34
|
Xie J, Gorlé N, Vandendriessche C, Van Imschoot G, Van Wonterghem E, Van Cauwenberghe C, Parthoens E, Van Hamme E, Lippens S, Van Hoecke L, Vandenbroucke RE. Low-grade peripheral inflammation affects brain pathology in the App NL-G-Fmouse model of Alzheimer's disease. Acta Neuropathol Commun 2021; 9:163. [PMID: 34620254 PMCID: PMC8499584 DOI: 10.1186/s40478-021-01253-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022] Open
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disease characterized by the accumulation of amyloid β (Aβ) and neurofibrillary tangles. The last decade, it became increasingly clear that neuroinflammation plays a key role in both the initiation and progression of AD. Moreover, also the presence of peripheral inflammation has been extensively documented. However, it is still ambiguous whether this observed inflammation is cause or consequence of AD pathogenesis. Recently, this has been studied using amyloid precursor protein (APP) overexpression mouse models of AD. However, the findings might be confounded by APP-overexpression artifacts. Here, we investigated the effect of low-grade peripheral inflammation in the APP knock-in (AppNL-G-F) mouse model. This revealed that low-grade peripheral inflammation affects (1) microglia characteristics, (2) blood-cerebrospinal fluid barrier integrity, (3) peripheral immune cell infiltration and (4) Aβ deposition in the brain. Next, we identified mechanisms that might cause this effect on AD pathology, more precisely Aβ efflux, persistent microglial activation and insufficient Aβ clearance, neuronal dysfunction and promotion of Aβ aggregation. Our results further strengthen the believe that even low-grade peripheral inflammation has detrimental effects on AD progression and may further reinforce the idea to modulate peripheral inflammation as a therapeutic strategy for AD.![]()
Collapse
|
35
|
Chen L, Zhou Y, Wang J, Li K, Zhang Z, Peng M. The adenosine A 2A receptor alleviates postoperative delirium-like behaviors by restoring blood cerebrospinal barrier permeability in rats. J Neurochem 2021; 158:980-996. [PMID: 34033116 DOI: 10.1111/jnc.15436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/04/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022]
Abstract
Postoperative delirium (POD) is a common post-operative complication in elderly patients that is associated with increased morbidity and mortality. However, the neuropathogenesis of this complication remains unknown. The blood-cerebrospinal fluid barrier (BCB) and brain-blood barrier (BBB) are composed of tight junctions between cells that form physical barriers, and BBB damage plays an important role in the neuropathogenesis of POD. Nevertheless, the role of BCB in POD remains to be elucidated. Herein, we investigated the effect of adenosine A2A receptor (A2A R), a key regulator of the permeability of barriers, on surgery-induced increased permeability of BCB and POD-like behaviors. Open field, buried food, and Y maze tests were used to evaluate behavioral changes in rats after surgery. Levels of tight junction proteins, adherens junction proteins, A2A R, GTP-RhoA, and ROCK2 in the choroid plexus were assessed by western blotting. The concentrations of NaFI and FITC-dextran in the cerebrospinal fluid (CSF) were detected by fluorescence spectrophotometry. Transmission electron microscopy was applied to observe the ultrastructure of the choroid plexus. Surgery/anesthesia decreased the levels of tight junction (e.g., ZO-1, occludin, and claudin1) proteins, increased concentrations of NaFI and FITC-dextran in CSF, damaged the ultrastructure of choroid plexus, and induced POD-like behaviors in rats. An A2A R antagonist alleviated POD-like behaviors in rats. Furthermore, the A2A R antagonist increased the levels of tight junction proteins and restored the permeability of BCB in rats with POD. Fasudil, a selective Rho-associated protein kinase 2 (ROCK2) inhibitor, ameliorated POD-like behaviors induced by A2A R activation. Moreover, fasudil also abolished the increased levels of GTP-RhoA/ROCK2, decreased levels of tight junction proteins, and increased permeability of BCB caused by A2A R activation. Our findings demonstrate that A2A R might participate in regulating BCB permeability in rats with POD via the RhoA/ROCK2 signaling pathway, which suggests the potential of A2A R as a therapeutic target for POD.
Collapse
Affiliation(s)
- Lei Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Zhou
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiayu Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ke Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mian Peng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
36
|
Marizzoni M, Cattaneo A, Mirabelli P, Festari C, Lopizzo N, Nicolosi V, Mombelli E, Mazzelli M, Luongo D, Naviglio D, Coppola L, Salvatore M, Frisoni GB. Short-Chain Fatty Acids and Lipopolysaccharide as Mediators Between Gut Dysbiosis and Amyloid Pathology in Alzheimer's Disease. J Alzheimers Dis 2021; 78:683-697. [PMID: 33074224 DOI: 10.3233/jad-200306] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Metagenomic data support an association between certain bacterial strains and Alzheimer's disease (AD), but their functional dynamics remain elusive. OBJECTIVE To investigate the association between amyloid pathology, bacterial products such as lipopolysaccharide (LPS) and short chain fatty acids (SCFAs: acetate, valerate, butyrate), inflammatory mediators, and markers of endothelial dysfunction in AD. METHODS Eighty-nine older persons with cognitive performance from normal to dementia underwent florbetapir amyloid PET and blood collection. Brain amyloidosis was measured with standardized uptake value ratio versus cerebellum. Blood levels of LPS were measured by ELISA, SCFAs by mass spectrometry, cytokines by using real-time PCR, and biomarkers of endothelial dysfunction by flow cytometry. We investigated the association between the variables listed above with Spearman's rank test. RESULTS Amyloid SUVR uptake was positively associated with blood LPS (rho≥0.32, p≤0.006), acetate and valerate (rho≥0.45, p < 0.001), pro-inflammatory cytokines (rho≥0.25, p≤0.012), and biomarkers of endothelial dysfunction (rho≥0.25, p≤0.042). In contrast, it was negatively correlated with butyrate (rho≤-0.42, p≤0.020) and the anti-inflammatory cytokine IL10 (rho≤-0.26, p≤0.009). Endothelial dysfunction was positively associated with pro-inflammatory cytokines, acetate and valerate (rho≥0.25, p≤0.045) and negatively with butyrate and IL10 levels (rho≤-0.25, p≤0.038). CONCLUSION We report a novel association between gut microbiota-related products and systemic inflammation with brain amyloidosis via endothelial dysfunction, suggesting that SCFAs and LPS represent candidate pathophysiologic links between the gut microbiota and AD pathology.
Collapse
Affiliation(s)
- Moira Marizzoni
- Laboratory of Neuroimaging and Alzheimer's Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Annamaria Cattaneo
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Cristina Festari
- Laboratory of Neuroimaging and Alzheimer's Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Nicola Lopizzo
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Valentina Nicolosi
- Laboratory of Neuroimaging and Alzheimer's Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Elisa Mombelli
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Monica Mazzelli
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Daniele Naviglio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, Naples, Italy
| | | | | | - Giovanni B Frisoni
- Memory Clinic and LANVIE - Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, Geneva, Switzerland
| |
Collapse
|
37
|
Zhang X, Yuan H, Lyu J, Meng X, Tian Q, Li Y, Zhang J, Xu X, Su J, Hou H, Li D, Sun B, Wang W, Wang Y. Association of dementia with immunoglobulin G N-glycans in a Chinese Han Population. NPJ Aging Mech Dis 2021; 7:3. [PMID: 33542243 PMCID: PMC7862610 DOI: 10.1038/s41514-021-00055-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 01/06/2021] [Indexed: 12/24/2022] Open
Abstract
Immunoglobulin G (IgG) functionality can drastically change from anti- to proinflammatory by alterations in the IgG N-glycan patterns. Our previous studies have demonstrated that IgG N-glycans associated with the risk factors of dementia, such as aging, dyslipidemia, type 2 diabetes mellitus, hypertension, and ischemic stroke. Therefore, the aim is to investigate whether the effects of IgG N-glycan profiles on dementia exists in a Chinese Han population. A case–control study, including 81 patients with dementia, 81 age- and gender-matched controls with normal cognitive functioning (NC) and 108 non-matched controls with mild cognitive impairment (MCI) was performed. Plasma IgG N-glycans were separated by ultra-performance liquid chromatography. Fourteen glycan peaks reflecting decreased of sialylation and core fucosylation, and increased bisecting N-acetylglucosamine (GlcNAc) N-glycan structures were of statistically significant differences between dementia and NC groups after controlling for confounders (p < 0.05; q < 0.05). Similarly, the differences for these 14 initial glycans were statistically significant between AD and NC groups after adjusting for the effects of confounders (p < 0.05; q < 0.05). The area under the receiver operating curve (AUC) value of the model consisting of GP8, GP9, and GP14 was determined to distinguish dementia from NC group as 0.876 [95% confidence interval (CI): 0.815–0.923] and distinguish AD from NC group as 0.887 (95% CI: 0.819–0.936). Patients with dementia were of an elevated proinflammatory activity via the significant changes of IgG glycome. Therefore, IgG N-glycans might contribute to be potential novel biomarkers for the neurodegenerative process risk assessment of dementia.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, 100069, China.,Department of Anesthesiology, Sanbo Brain Hospital, Capital Medical University, Beijing, 100095, China
| | - Hui Yuan
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, 271000, China
| | - Jihui Lyu
- Center for Cognitive Disorders, Beijing Geriatric Hospital, Beijing, 100095, China
| | - Xiaoni Meng
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, 100069, China
| | - Qiuyue Tian
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, 100069, China
| | - Yuejin Li
- School of public health, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271000, China
| | - Jie Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, 100069, China
| | - Xizhu Xu
- School of public health, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271000, China
| | - Jing Su
- Department of Geriatrics, Tai'an City Central Hospital, Tai'an, 271000, China
| | - Haifeng Hou
- School of public health, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271000, China
| | - Dong Li
- School of public health, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271000, China
| | - Baoliang Sun
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271000, China
| | - Wei Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, 100069, China. .,School of public health, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, 271000, China. .,School of Medical and Health Sciences, Edith Cowan University, Perth, WA, 6027, Australia.
| | - Youxin Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, 100069, China. .,School of Medical and Health Sciences, Edith Cowan University, Perth, WA, 6027, Australia.
| |
Collapse
|
38
|
Cullen NC, Mälarstig AN, Stomrud E, Hansson O, Mattsson-Carlgren N. Accelerated inflammatory aging in Alzheimer's disease and its relation to amyloid, tau, and cognition. Sci Rep 2021; 11:1965. [PMID: 33479445 PMCID: PMC7820414 DOI: 10.1038/s41598-021-81705-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022] Open
Abstract
It is unclear how pathological aging of the inflammatory system relates to Alzheimer's disease (AD). We tested whether age-related inflammatory changes in cerebrospinal fluid (CSF) and plasma exist across different stages of AD, and whether such changes related to AD pathology. Linear regression was first used model chronological age in amyloid-β negative, cognitively unimpaired individuals (Aβ- CU; n = 312) based on a collection of 73 inflammatory proteins measured in both CSF and plasma. Fitted models were then applied on protein levels from Aβ+ individuals with mild cognitive impairment (Aβ+ MCI; n = 150) or Alzheimer's disease dementia (Aβ+ AD; n = 139) to test whether the age predicted from proteins alone ("inflammatory age") differed significantly from true chronological age. Aβ- individuals with subjective cognitive decline (Aβ- SCD; n = 125) or MCI (Aβ- MCI; n = 104) were used as an independent contrast group. The difference between inflammatory age and chronological age (InflammAGE score) was then assessed in relation to core AD biomarkers of amyloid, tau, and cognition. Both CSF and plasma inflammatory proteins were significantly associated with age in Aβ- CU individuals, with CSF-based proteins predicting chronological age better than plasma-based counterparts. Meanwhile, the Aβ- SCD and validation Aβ- CU groups were not characterized by significant inflammatory aging, while there was increased inflammatory aging in Aβ- MCI patients for CSF but not plasma inflammatory markers. Both CSF and plasma inflammatory changes were seen in the Aβ+ MCI and Aβ+ AD groups, with varying degrees of change compared to Aβ- CU and Aβ- SCD groups. Finally, CSF inflammatory changes were highly correlated with amyloid, tau, general neurodegeneration, and cognition, while plasma changes were mostly associated with amyloid and cognition. Inflammatory pathways change during aging and are specifically altered in AD, tracking closely with pathological hallmarks. These results have implications for tracking AD progression and for suggesting possible pathways for drug targeting.
Collapse
Affiliation(s)
- Nicholas C Cullen
- Clinical Memory Research Unit, Department of Clinical Sciences, Faculty of Medicine, Lund University, Sölvegatan 19, BMC - C11, 223 62, Lund, Sweden.
| | - A Nders Mälarstig
- Pfizer Worldwide Research & Development, Stockholm, Sweden
- Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Faculty of Medicine, Lund University, Sölvegatan 19, BMC - C11, 223 62, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Faculty of Medicine, Lund University, Sölvegatan 19, BMC - C11, 223 62, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Faculty of Medicine, Lund University, Sölvegatan 19, BMC - C11, 223 62, Lund, Sweden.
- Department of Neurology, Skåne University Hospital, Lund, Sweden.
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
39
|
Robbins J, Busquets O, Tong M, de la Monte SM. Dysregulation of Insulin-Linked Metabolic Pathways in Alzheimer's Disease: Co-Factor Role of Apolipoprotein E ɛ4. J Alzheimers Dis Rep 2020; 4:479-493. [PMID: 33344887 PMCID: PMC7739986 DOI: 10.3233/adr-200238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Brain insulin resistance and deficiency are well-recognized abnormalities in Alzheimer's disease (AD) and likely mediators of impaired energy metabolism. Since apolipoprotein E (APOE) is a major risk factor for late-onset AD, it was of interest to examine its potential contribution to altered insulin-linked signaling networks in the brain. OBJECTIVE The main goal was to evaluate the independent and interactive contributions of AD severity and APOE ɛ4 dose on brain expression of insulin-related polypeptides and inflammatory mediators of metabolic dysfunction. METHODS Postmortem fresh frozen frontal lobe tissue from banked cases with known APOE genotypes and different AD Braak stages were used to measure insulin network polypeptide immunoreactivity with a commercial multiplex enzyme-linked immunosorbent assay (ELISA). RESULTS Significant AD Braak stage and APOE genotype-related abnormalities in insulin, C-peptide, gastric inhibitory polypeptide (GIP), glucaton-like peptide-1 (GLP-1), leptin, ghrelin, glucagon, resistin, and plasminogen activator inhibitor-1 (PAI-1) were detected. The main factors inhibiting polypeptide expression and promoting neuro-inflammatory responses included AD Braak stage and APOE ɛ4/ɛ4 rather than ɛ3/ɛ4. CONCLUSION This study demonstrates an expanded role for impaired expression of insulin-related network polypeptides as well as neuroinflammatory mediators of brain insulin resistance in AD pathogenesis and progression. In addition, the findings show that APOE has independent and additive effects on these aberrations in brain polypeptide expression, but the impact is decidedly greater for APOE ɛ4/ɛ4 than ɛ3/ɛ4.
Collapse
Affiliation(s)
- James Robbins
- Alpert Medical School of Brown University, Providence, RI, USA
| | - Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Ming Tong
- Alpert Medical School of Brown University, Providence, RI, USA,Department of Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Suzanne M. de la Monte
- Alpert Medical School of Brown University, Providence, RI, USA,Department of Medicine, Rhode Island Hospital, Providence, RI, USA,Departments of Pathology and Laboratory Medicine Providence VA Medical Center, Rhode Island Hospital, and the Women and Infants Hospital of Rhode Island, Providence, RI, USA,Correspondence to: Dr. Suzanne M. de la Monte, MD, MPH, Rhode Island Hospital, 55 Claverick Street, Room 419, Providence, RI 02903, USA. Tel.: +1 401 444 7364; Fax: +1 401 444 2939; E-mail:
| |
Collapse
|
40
|
Wu CY, Bawa KK, Ouk M, Leung N, Yu D, Lanctôt KL, Herrmann N, Pakosh M, Swardfager W. Neutrophil activation in Alzheimer's disease and mild cognitive impairment: A systematic review and meta-analysis of protein markers in blood and cerebrospinal fluid. Ageing Res Rev 2020; 62:101130. [PMID: 32712109 DOI: 10.1016/j.arr.2020.101130] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/28/2020] [Accepted: 07/20/2020] [Indexed: 12/29/2022]
Abstract
Inflammation is involved in the pathophysiology of Alzheimer's disease (AD), with multiple inflammatory processes implicated in its risk and progression. This review included original peer-reviewed studies measuring the cerebrospinal fluid or peripheral blood concentrations of protein markers specifically related to neutrophil activity in healthy controls (HC) and in patients with AD or mild cognitive impairment (MCI). A total of 35 studies (NHC = 3095, NAD = 2596, NMCI = 1203) were included. Random-effects meta-analyses were used to estimate between-groups standardized mean differences (SMD) and 95 % confidence intervals. In blood, concentrations of myeloperoxidase (MPO; NAD/NHC = 271/209, SMD = 0.41 [0.20, 0.62]; I2 = 15.7 %) and neutrophil gelatinase associated lipocalin (NGAL; NAD/NHC = 273/185, SMD = 0.30 [0.11, 0.49]; I2 < 0.005 %) were significantly higher in AD relative to HC. Peripheral blood concentrations of NGAL were also higher in MCI compared to HC (NMCI/NHC = 489/145, SMD = 0.39 [0.11, 0.67]; I2 = 38.6 %). None of the protein markers exhibited a significant difference between HC, MCI, or AD groups in the cerebrospinal fluid. The evidence suggests that peripheral neutrophil activation, as indicated by blood concentrations of NGAL and MPO, may be a pathological feature of cognitive impairment due to AD, evident at stages of MCI and AD dementia.
Collapse
Affiliation(s)
- Che-Yuan Wu
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Kritleen K Bawa
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Michael Ouk
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Nathan Leung
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Di Yu
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Krista L Lanctôt
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada; Department of Psychiatry, Faculty of Medicine, University of Toronto, 250 College Street, 8th Floor, Toronto, ON, M5T 1R8, Canada; KITE UHN Toronto Rehabilitation Institute, 347 Rumsey Rd, East York, ON, M4G 2V6, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Nathan Herrmann
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada; Department of Psychiatry, Faculty of Medicine, University of Toronto, 250 College Street, 8th Floor, Toronto, ON, M5T 1R8, Canada
| | - Maureen Pakosh
- KITE UHN Toronto Rehabilitation Institute, 347 Rumsey Rd, East York, ON, M4G 2V6, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada; Hurvitz Brain Sciences Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada; KITE UHN Toronto Rehabilitation Institute, 347 Rumsey Rd, East York, ON, M4G 2V6, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada.
| |
Collapse
|
41
|
Wissler Gerdes EO, Zhu Y, Weigand BM, Tripathi U, Burns TC, Tchkonia T, Kirkland JL. Cellular senescence in aging and age-related diseases: Implications for neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:203-234. [PMID: 32854855 PMCID: PMC7656525 DOI: 10.1016/bs.irn.2020.03.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging is the major predictor for developing multiple neurodegenerative diseases, including Alzheimer's disease (AD) other dementias, and Parkinson's disease (PD). Senescent cells, which can drive aging phenotypes, accumulate at etiological sites of many age-related chronic diseases. These cells are resistant to apoptosis and can cause local and systemic dysfunction. Decreasing senescent cell abundance using senolytic drugs, agents that selectively target these cells, alleviates neurodegenerative diseases in preclinical models. In this review, we consider roles of senescent cells in neurodegenerative diseases and potential implications of senolytic agents as an innovative treatment.
Collapse
Affiliation(s)
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - B Melanie Weigand
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Utkarsh Tripathi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Terence C Burns
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
42
|
Söderbom G, Zeng BY. The NLRP3 inflammasome as a bridge between neuro-inflammation in metabolic and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:345-391. [PMID: 32739011 DOI: 10.1016/bs.irn.2020.03.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Evidence increasingly suggests that type 2 diabetes mellitus (T2DM) is a risk factor for neurodegenerative diseases (NDDs), such as Alzheimer's disease (AD) and Parkinson's disease (PD). These diseases share many pathological processes, including oxidative stress, local inflammation/neuroinflammation and chronic, low-grade (systemic) inflammation, which are exacerbated by aging, a common risk factor for T2DM and NDDs. Here, we focus on the link between chronic inflammation driven by peripheral metabolic disease and how this may impact neurodegeneration in AD and PD. We review the relationship between these common pathological processes in AD and PD from the perspective of the "pro-inflammatory" signaling of the nucleotide-binding oligomerization domain (NOD)-, leucine-rich repeat- (LRR)-, and pyrin domain-containing protein 3 (NLRP3) inflammasome complex. Since the need for effective disease-modifying therapies in T2DM, AD and PD is significant, the relationship between these diseases is important as a positive clinical impact on one may benefit the others. We briefly consider how novel strategies may target neuro-inflammation and provide potential therapies for AD and PD.
Collapse
Affiliation(s)
| | - Bai-Yun Zeng
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
43
|
Adenosine and Metabotropic Glutamate Receptors Are Present in Blood Serum and Exosomes from SAMP8 Mice: Modulation by Aging and Resveratrol. Cells 2020; 9:cells9071628. [PMID: 32645849 PMCID: PMC7407497 DOI: 10.3390/cells9071628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/27/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022] Open
Abstract
Adenosine (ARs) and metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors (GPCRs) that are modulated in the brain of SAMP8 mice, an animal model of Alzheimer's disease (AD). In the present work, it is shown the presence of ARs and mGluRs in blood serum and derived exosomes from SAMP8 mice as well as its possible modulation by aging and resveratrol (RSV) consumption. In blood serum, adenosine A1 and A2A receptors remained unaltered from 5 to 7 months of age. However, an age-related decrease in adenosine level was observed, while 5'-Nucleotidase activity was not modulated. Regarding the glutamatergic system, it was observed a decrease in mGluR5 density and glutamate levels in older mice. In addition, dietary RSV supplementation caused an age-dependent modulation in both adenosinergic and glutamatergic systems. These GPCRs were also found in blood serum-derived exosomes, which might suggest that these receptors could be released into circulation via exosomes. Interestingly, changes elicited by age and RSV supplementation on mGluR5 density, and adenosine and glutamate levels were similar to that detected in whole-brain. Therefore, we might suggest that the quantification of these receptors, and their corresponding endogenous ligands, in blood serum could have predictive value for early diagnosis in combination with other distinctive hallmarks of AD.
Collapse
|
44
|
Liu X, Chen J, Geng R, Wei R, Xu P, Chen B, Liu K, Yang L. Sex- and age-specific mild cognitive impairment is associated with low hand grip strength in an older Chinese cohort. J Int Med Res 2020; 48:300060520933051. [PMID: 32602799 PMCID: PMC7328492 DOI: 10.1177/0300060520933051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Few studies have demonstrated the impact of characteristics like age and sex on the association between hand grip strength (HGS) and mild cognitive impairment (MCI). In this cross-sectional study, we aimed to examine the effects of sex and age on the relationship between HGS and MCI. Methods We enrolled older adults age ≥60 years (n = 1009) and measured HGS and MCI in all participants. We analyzed the differences in MCI prevalence among the different variables. The role of sex and age in the association between MCI and HGS was analyzed using binary logistic regression. Results Women had significantly higher prevalence of MCI than men, as did the older group (age ≥70 years) compared with the younger group (age 60–70 years). In men, the low and middle HGS tertiles were significantly associated with MCI. In contrast, only the low tertile of HGS was associated with MCI in women. In the older group, the low tertile of HGS was significantly associated with MCI, which was not observed in the younger group. Conclusions HGS was associated with MCI in older adults, and this association was stronger in men. HGS may be useful for evaluating MCI in older adults.
Collapse
Affiliation(s)
- Xinji Liu
- Nutritional Department of Anhui No. 2 Provincial People's Hospital, Hefei, Anhui, China.,School of Public Health, Department of Nutrition and Food Hygiene, Anhui Medical University, Anhui Hefei, China
| | - Jun Chen
- School of Public Health, Southern Medical University, Guangdong Guangzhou, China
| | - Renwen Geng
- School of Public Health, Southern Medical University, Guangdong Guangzhou, China
| | - Rong Wei
- School of Public Health, Department of Nutrition and Food Hygiene, Anhui Medical University, Anhui Hefei, China
| | - Peiru Xu
- School of Public Health, Department of Epidemiology and Health Statistics, Anhui Medical University, Anhui Hefei, China
| | - Beijing Chen
- School of Public Health, Department of Nutrition and Food Hygiene, Anhui Medical University, Anhui Hefei, China
| | - Kaiyong Liu
- School of Public Health, Department of Nutrition and Food Hygiene, Anhui Medical University, Anhui Hefei, China
| | - Linsheng Yang
- School of Public Health, Department of Epidemiology and Health Statistics, Anhui Medical University, Anhui Hefei, China
| |
Collapse
|
45
|
Shen XN, Lu Y, Tan CTY, Liu LY, Yu JT, Feng L, Larbi A. Identification of inflammatory and vascular markers associated with mild cognitive impairment. Aging (Albany NY) 2020; 11:2403-2419. [PMID: 31039131 PMCID: PMC6520012 DOI: 10.18632/aging.101924] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/24/2019] [Indexed: 12/27/2022]
Abstract
Biochemical processes have been associated with the pathogenesis of mild cognitive impairment (MCI) and dementia, including chronic inflammation, dysregulation of membrane lipids and disruption of neurotransmitter pathways. However, research investigating biomarkers of these processes in MCI remained sparse and inconsistent. To collect fresh evidence, we evaluated the performance of several potential markers in a cohort of 57 MCI patients and 57 cognitively healthy controls. MCI patients showed obviously increased levels of plasma TNF-α (p = 0.045) and C-peptide (p = 0.004) as well as decreased levels of VEGF-A (p = 0.042) and PAI-1 (p = 0.019), compared with controls. In addition, our study detected significant correlations of plasma sTNFR-1 (MCI + Control: B = -6.529, p = 0.020; MCI: B = -9.865, p = 0.011) and sIL-2Rα (MCI + Control: B = -7.010, p = 0.007; MCI: B = -11.834, p = 0.003) levels with MoCA scores in the whole cohort and the MCI group. These findings corroborate the inflammatory and vascular hypothesis for dementia. Future studies are warranted to determine their potential as early biomarkers for cognitive deficits and explore the related mechanisms.
Collapse
Affiliation(s)
- Xue-Ning Shen
- Department of Neurology, Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanxia Lu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | - Crystal Tze Ying Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | - Ling-Yun Liu
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Neurology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology, Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Feng
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biology, Faculty of Sciences, University Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
46
|
Martins GL, Duarte RCF, Mukhamedyarov MA, Palotás A, Ferreira CN, Reis HJ. Inflammatory and Infectious Processes Serve as Links between Atrial Fibrillation and Alzheimer's Disease. Int J Mol Sci 2020; 21:E3226. [PMID: 32370194 PMCID: PMC7247326 DOI: 10.3390/ijms21093226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 04/25/2020] [Accepted: 04/30/2020] [Indexed: 11/16/2022] Open
Abstract
Atrial fibrillation (AF) is one of the most prevalent forms of arrhythmia that carries an increased risk of stroke which, in turn, is strongly associated with cognitive decline. The majority of dementia cases are caused by Alzheimer's disease (AD) with obscure pathogenesis. While the exact mechanisms are unknown, the role of inflammatory processes and infectious agents have recently been implicated in both AD and AF, suggesting a common link between these maladies. Here, we present the main shared pathways underlying arrhythmia and memory loss. The overlapping predictive biomarkers and emerging joint pharmacological approaches are also discussed.
Collapse
Affiliation(s)
- Gabriela Lopes Martins
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, BR-31270-901 Belo Horizonte, Brazil; (G.L.M.); (R.C.F.D.); (C.N.F.); (H.J.R.)
| | - Rita Carolina Figueiredo Duarte
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, BR-31270-901 Belo Horizonte, Brazil; (G.L.M.); (R.C.F.D.); (C.N.F.); (H.J.R.)
| | | | - András Palotás
- Asklepios-Med (Private Medical Practice and Research Center), H-6722 Szeged, Hungary
- Institute of Fundamental Medicine and Biology, Kazan Federal University, R-420008 Kazan, Russia
| | - Cláudia Natália Ferreira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, BR-31270-901 Belo Horizonte, Brazil; (G.L.M.); (R.C.F.D.); (C.N.F.); (H.J.R.)
| | - Helton José Reis
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, BR-31270-901 Belo Horizonte, Brazil; (G.L.M.); (R.C.F.D.); (C.N.F.); (H.J.R.)
| |
Collapse
|
47
|
Martínez-Cué C, Rueda N. Cellular Senescence in Neurodegenerative Diseases. Front Cell Neurosci 2020; 14:16. [PMID: 32116562 PMCID: PMC7026683 DOI: 10.3389/fncel.2020.00016] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a homeostatic biological process characterized by a permanent state of cell cycle arrest that can contribute to the decline of the regenerative potential and function of tissues. The increased presence of senescent cells in different neurodegenerative diseases suggests the contribution of senescence in the pathophysiology of these disorders. Although several factors can induce senescence, DNA damage, oxidative stress, neuroinflammation, and altered proteostasis have been shown to play a role in its onset. Oxidative stress contributes to accelerated aging and cognitive dysfunction stages affecting neurogenesis, neuronal differentiation, connectivity, and survival. During later life stages, it is implicated in the progression of cognitive decline, synapse loss, and neuronal degeneration. Also, neuroinflammation exacerbates oxidative stress, synaptic dysfunction, and neuronal death through the harmful effects of pro-inflammatory cytokines on cell proliferation and maturation. Both oxidative stress and neuroinflammation can induce DNA damage and alterations in DNA repair that, in turn, can exacerbate them. Another important feature associated with senescence is altered proteostasis. Because of the disruption in the function and balance of the proteome, senescence can modify the proper synthesis, folding, quality control, and degradation rate of proteins producing, in some diseases, misfolded proteins or aggregation of abnormal proteins. There is an extensive body of literature that associates cellular senescence with several neurodegenerative disorders including Alzheimer’s disease (AD), Down syndrome (DS), and Parkinson’s disease (PD). This review summarizes the evidence of the shared neuropathological events in these neurodegenerative diseases and the implication of cellular senescence in their onset or aggravation. Understanding the role that cellular senescence plays in them could help to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| |
Collapse
|
48
|
Nakagawa Y, Yamada S. Metal homeostasis disturbances in neurodegenerative disorders, with special emphasis on Creutzfeldt-Jakob disease - Potential pathogenetic mechanism and therapeutic implications. Pharmacol Ther 2019; 207:107455. [PMID: 31863817 DOI: 10.1016/j.pharmthera.2019.107455] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
Creutzfeldt-Jakob disease (CJD) is characterized by a rapidly progressive dementia often accompanied by myoclonus and other signs of brain dysfunction, relying on the conversion of the normal cellular form of the prion protein (PrPC) to a misfolded form (PrPSc). The neuropathological changes include spongiform degeneration, neuronal loss, astrogliosis, and deposition of PrPSc. It is still unclear how these pathological changes correlate with the development of CJD symptoms because few patients survive beyond 2 years after diagnosis. Inasmuch as the symptoms of CJD overlap some of those observed in Alzheimer's, Parkinson's, and Huntington's diseases, there may be some underlying pathologic mechanisms associated with CJD that may contribute to the symptoms of non-prion neurodegenerative diseases as well. Data suggest that imbalance of metals, including copper, zinc, iron, and manganese, induces abnormalities in processing and degradation of prion proteins that are accompanied by self-propagation of PrPSc. These events appear to be responsible for glutamatergic synaptic dysfunctions, neuronal death, and PrPSc aggregation. Given that the prodromal symptoms of CJD such as sleep disturbances and mood disorders are associated with brain stem and limbic system dysfunction, the pathological changes may initially occur in these brain regions, then spread throughout the entire brain. Alterations in cerebrospinal fluid homeostasis, which may be linked to imbalance of these metals, seem to be more important than neuroinflammation in causing the cell death. It is proposed that metal dyshomeostasis could be responsible for the initiation and progression of the pathological changes associated with symptoms of CJD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
49
|
Calderón-Garcidueñas L, Mukherjee PS, Waniek K, Holzer M, Chao CK, Thompson C, Ruiz-Ramos R, Calderón-Garcidueñas A, Franco-Lira M, Reynoso-Robles R, Gónzalez-Maciel A, Lachmann I. Non-Phosphorylated Tau in Cerebrospinal Fluid is a Marker of Alzheimer's Disease Continuum in Young Urbanites Exposed to Air Pollution. J Alzheimers Dis 2019; 66:1437-1451. [PMID: 30412505 DOI: 10.3233/jad-180853] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Long-term exposure to fine particulate matter (PM2.5) and ozone (O3) above USEPA standards is associated with Alzheimer's disease (AD) risk. Metropolitan Mexico City (MMC) children exhibit subcortical pretangles in infancy and cortical tau pre-tangles, NFTs, and amyloid phases 1-2 by the 2nd decade. Given their AD continuum, we measured in 507 normal cerebrospinal fluid (CSF) samples (MMC 354, controls 153, 12.82±6.73 y), a high affinity monoclonal non-phosphorylated tau antibody (non-P-Tau), as a potential biomarker of AD and axonal damage. In 81 samples, we also measured total tau (T-Tau), tau phosphorylated at threonine 181 (P-Tau), amyloid-β1-42, BDNF, and vitamin D. We documented by electron microscopy myelinated axonal size and the pathology associated with combustion-derived nanoparticles (CDNPs) in anterior cingulate cortex white matter in 6 young residents (16.25±3.34 y). Non-P-Tau showed a strong increase with age significantly faster among MMC versus controls (p = 0.0055). Aβ1 - 42 and BDNF concentrations were lower in MMC children (p = 0.002 and 0.03, respectively). Anterior cingulate cortex showed a significant decrease (p = <0.0001) in the average axonal size and CDNPs were associated with organelle pathology. Significant age increases in non-P-Tau support tau changes early in a population with axonal pathology and evolving AD hallmarks in the first two decades of life. Non-P-Tau is an early biomarker of axonal damage and potentially valuable to monitor progressive longitudinal changes along with AD multianalyte classical CSF markers. Neuroprotection of young urbanites with PM2.5 and CDNPs exposures ought to be a public health priority to halt the development of AD in the first two decades of life.
Collapse
Affiliation(s)
| | | | | | - Max Holzer
- Paul-Flechsig-Institute for Brain Research, Leipzig, Germany
| | | | | | - Rubén Ruiz-Ramos
- Instituto de Medicina Forense, Universidad Veracruzana, Boca del Rio, Mexico
| | | | | | | | | | | |
Collapse
|
50
|
Gofrit ON, Klein BY, Cohen IR, Ben-Hur T, Greenblatt CL, Bercovier H. Bacillus Calmette-Guérin (BCG) therapy lowers the incidence of Alzheimer's disease in bladder cancer patients. PLoS One 2019; 14:e0224433. [PMID: 31697701 PMCID: PMC6837488 DOI: 10.1371/journal.pone.0224433] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/14/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) affects one in ten people older than 65 years. Thus far, there is no cure or even disease-modifying treatment for this disease. The immune system is a major player in the pathogenesis of AD. Bacillus Calmette-Guérin (BCG), developed as a vaccine against tuberculosis, modulates the immune system and reduces recurrence of non-muscle invasive bladder cancer. Theoretical considerations suggested that treatment with BCG may decrease the risk of AD. We tested this hypothesis on a natural population of bladder cancer patients. METHODS AND FINDINGS After removing all bladder cancer patients presenting with AD or developing AD within one-year following diagnosis of bladder cancer, we collected data on a total of 1371 patients (1134 males and 237 females) who were followed for at least one year after the diagnosis of bladder cancer. The mean age at diagnosis of bladder cancer was 68.1 years (SD 13.0). Adjuvant post-operative intra-vesical treatment with BCG was given to 878 (64%) of these patients. The median period post-operative follow-up was 8 years. During follow-up, 65 patients developed AD at a mean age of 84 years (SD 5.9), including 21 patients (2.4%) who had been treated with BCG and 44 patients (8.9%) who had not received BCG. Patients who had been treated with BCG manifested more than 4-fold less risk for AD than those not treated with BCG. The Cox proportional hazards regression model and the Kaplan-Meier analysis of AD free survival both indicated high significance: patients not treated with BCG had a significantly higher risk of developing AD compared to BCG treated patients (HR 4.778, 95%CI: 2.837-8.046, p = 4.08x10-9 and Log Rank Chi-square 42.438, df = 1, p = 7.30x10-11, respectively). Exposure to BCG did not modify the prevalence of Parkinson's disease, 1.9% in BCG treated patients and 1.6% in untreated (Fisher's Exact Test, p = 1). CONCLUSIONS Bladder cancer patients treated with BCG were significantly less likely to develop AD at any age than patients who were not so treated. This finding of a retrospective study suggests that BCG treatment might also reduce the incidence of AD in the general population. Confirmation of such effects of BCG in other retrospective studies would support prospective studies of BCG in AD.
Collapse
Affiliation(s)
- Ofer N. Gofrit
- Department of Urology, Hadassah- Hebrew University Medical Center, Jerusalem, Israel
- * E-mail: (HB); (ONG)
| | - Benjamin Y. Klein
- Department of Microbiology and Molecular Genetics, Hebrew University Jerusalem, Israel
| | - Irun R. Cohen
- Department of Immunology, Weizmann Institute, Rehovot, Israel
| | - Tamir Ben-Hur
- Department of Neurology Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Charles L. Greenblatt
- Department of Microbiology and Molecular Genetics, Hebrew University Jerusalem, Israel
| | - Hervé Bercovier
- Department of Microbiology and Molecular Genetics, Hebrew University Jerusalem, Israel
- * E-mail: (HB); (ONG)
| |
Collapse
|