101
|
Liraglutide Has Anti-Inflammatory and Anti-Amyloid Properties in Streptozotocin-Induced and 5xFAD Mouse Models of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22020860. [PMID: 33467075 PMCID: PMC7829894 DOI: 10.3390/ijms22020860] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/07/2021] [Accepted: 01/14/2021] [Indexed: 12/22/2022] Open
Abstract
Recent clinical and epidemiological studies support the contention that diabetes mellitus (DM) is a strong risk factor for the development of Alzheimer’s disease (AD). The use of insulin cell toxin, streptozotocin (STZ), when injected into the lateral ventricles, develops an insulin resistant brain state (IRBS) and represents a non-transgenic, or sporadic AD model (SAD), with several AD-like neuropathological features. The present study explored the effects of an anti-diabetic drug, liraglutide (LIR), in reversing major pathological hallmarks in the prodromal disease stage of both the 5xFAD transgenic and SAD mouse models of AD. Three-month-old 5xFAD and age-matched wild type mice were given a single intracerebroventricular (i.c.v) injection of STZ or vehicle (saline) and were subsequently treated with LIR, intraperitoneally (IP), once a day for 30 days. The extent of neurodegeneration, Aβ plaque load, and key proteins associated with the insulin signaling pathways were measured using Western blot and neuroinflammation (via immunohistological assays) in the cortical and hippocampal regions of the brain were assessed following a series of behavioral tests used to measure cognitive function after LIR or vehicle treatments. Our results indicated that STZ significantly increased neuroinflammation, Aβ plaque deposition and disrupted insulin signaling pathway, while 25 nmol/kg LIR, when injected IP, significantly decreased neuroinflammatory responses in both SAD and 5xFAD mice before significant cognitive changes were observed, suggesting LIR can reduce early neuropathology markers prior to the emergence of overt memory deficits. Our results indicate that LIR has neuroprotective effects and has the potential to serve as an anti-inflammatory and anti-amyloid prophylactic therapy in the prodromal stages of AD.
Collapse
|
102
|
Johnson TS, Xiang S, Dong T, Huang Z, Cheng M, Wang T, Yang K, Ni D, Huang K, Zhang J. Combinatorial analyses reveal cellular composition changes have different impacts on transcriptomic changes of cell type specific genes in Alzheimer's Disease. Sci Rep 2021; 11:353. [PMID: 33432017 PMCID: PMC7801680 DOI: 10.1038/s41598-020-79740-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 12/09/2020] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease (AD) brains are characterized by progressive neuron loss and gliosis. Previous studies of gene expression using bulk tissue samples often fail to consider changes in cell-type composition when comparing AD versus control, which can lead to differences in expression levels that are not due to transcriptional regulation. We mined five large transcriptomic AD datasets for conserved gene co-expression module, then analyzed differential expression and differential co-expression within the modules between AD samples and controls. We performed cell-type deconvolution analysis to determine whether the observed differential expression was due to changes in cell-type proportions in the samples or to transcriptional regulation. Our findings were validated using four additional datasets. We discovered that the increased expression of microglia modules in the AD samples can be explained by increased microglia proportions in the AD samples. In contrast, decreased expression and perturbed co-expression within neuron modules in the AD samples was likely due in part to altered regulation of neuronal pathways. Several transcription factors that are differentially expressed in AD might account for such altered gene regulation. Similarly, changes in gene expression and co-expression within astrocyte modules could be attributed to combined effects of astrogliosis and astrocyte gene activation. Gene expression in the astrocyte modules was also strongly correlated with clinicopathological biomarkers. Through this work, we demonstrated that combinatorial analysis can delineate the origins of transcriptomic changes in bulk tissue data and shed light on key genes and pathways involved in AD.
Collapse
Affiliation(s)
- Travis S Johnson
- Department of Biostatistics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Shunian Xiang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Tianhan Dong
- Department of Pharmacology, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Zhi Huang
- Department of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Michael Cheng
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Tianfu Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Kai Yang
- Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Dong Ni
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen, 518060, China.
| | - Kun Huang
- Department of Medicine, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA.
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
103
|
Tsai ZR, Zhang HW, Tseng CH, Peng HC, Kok VC, Li GP, Hsiung CA, Hsu CY. Late-onset epilepsy and subsequent increased risk of dementia. Aging (Albany NY) 2021; 13:3573-3587. [PMID: 33429365 PMCID: PMC7906153 DOI: 10.18632/aging.202299] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Inflammation is considered as a key pathogenesis factor of dementia and epilepsy. However, epilepsy's association with dementia, particularly its role in the development of dementia, remains unclear. To evaluate the association between epilepsy and the risk of dementia, in Taiwan, we have now conducted a retrospective cohort study comprising 675 individuals (age, ≥50 years) with epilepsy and 2,025 matched control subjects without epilepsy. In order to match individuals diagnosed with epilepsy with those with no diagnosis of epilepsy (comparison cohort), we utilized exact matching at a ratio of 1:3. Compared with those in the comparison cohort, individuals in the epilepsy cohort had a significantly increased risk of developing dementia (adjusted hazard ratio = 2.87, p < 0.001). A similar result has been observed after stratifying for sex (adjusted hazard ratio in males = 2.95, p < 0.001; adjusted hazard ratio in females = 2.66, p < 0.001). To conclude, based on these data, epileptic individuals ≥50 years were at a greater risk of developing dementia than people who do not have epilepsy, which indicates that a diagnosis of epilepsy presents a greater risk for the development of dementia.
Collapse
Affiliation(s)
- Zhi-Ren Tsai
- Department of Computer Science and Information Engineering, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Taichung City Smart Transportation Big Data Research Center, Taichung, Taiwan
- Pervasive Artificial Intelligence Research (PAIR) Labs, Hsinchu, Taiwan
- Biomdcare Corporation, New Taipei, Taiwan
| | - Han-Wei Zhang
- Biomdcare Corporation, New Taipei, Taiwan
- Program for Aging, China Medical University, Taichung, Taiwan
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
- Institute of Electrical Control Engineering, Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Chun-Hung Tseng
- Department of Neurology, China Medical University Hospital, and School of Medicine, China Medical University, Taichung, Taiwan
| | | | - Victor C. Kok
- Disease Informatics Research Group, Asia University, Taichung, Taiwan
- Department of Internal Medicine, Kuang Tien General Hospital, Taichung, Taiwan
| | - Gao Ping Li
- Zhongshan Hospital, Affiliated Hospital of Fudan University, Shanghai, China
| | - Chao A. Hsiung
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Chun-Yi. Hsu
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
104
|
Siddiqui A, Akhtar S, Shah Z, Othman I, Kumari Y. Inflammation Drives Alzheimer's Disease: Emphasis on 5-lipoxygenase Pathways. Curr Neuropharmacol 2021; 19:885-895. [PMID: 32972344 PMCID: PMC8686299 DOI: 10.2174/1570159x18666200924122732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 11/22/2022] Open
Abstract
It is a known fact that inflammation affects several physiological processes, including the functioning of the central nervous system. Additionally, impairment of lipid mechanisms/pathways have been associated with a number of neurodegenerative disorders and Alzheimer's Disease (AD) is one of them. However, much attention has been given to the link between tau and beta- amyloid hypothesis in AD pathogenesis/prognosis. Increasing evidences suggest that biologically active lipid molecules could influence the pathophysiology of AD via a different mechanism of inflammation. This review intends to highlight the role of inflammatory responses in the context of AD with the emphasis on biochemical pathways of lipid metabolism enzyme, 5-lipoxygenase (5- LO).
Collapse
Affiliation(s)
- Aisha Siddiqui
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia
| | - Sayeed Akhtar
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha-21974, Kingdom of Saudi Arabia
| | - Zahoor Shah
- Department of Medicinal and Biological Chemistry, University of Toledo, 43614, 3000 Arlington Avenue, Toledo, Ohio, USA
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia
| | - Yatinesh Kumari
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Selangor, Malaysia
| |
Collapse
|
105
|
Zadik-Weiss L, Ritter S, Hermush V, Asher N, Avital A, Or R. Feline cognitive dysfunction as a model for Alzheimer's disease in the research of CBD as a potential treatment-a narrative review. J Cannabis Res 2020; 2:43. [PMID: 33526138 PMCID: PMC7819322 DOI: 10.1186/s42238-020-00054-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 11/13/2020] [Indexed: 11/15/2022] Open
Abstract
With the improvement in modern medicine, the world's human and feline (Felis catus, the domestic cat) population is aging. As the population grows older, there is an increase of age-related diseases, such as Alzheimer's disease in humans and feline cognitive dysfunction in felines, which shares many similarities with Alzheimer's disease. They both result in cognitive decline and lack effective treatments. In light of their pathological similarities, both occur at old age, and as domestic cats share the human environment and risk factors (cats are considered an indicator to the effect of environmental contaminants on humans as they share exposures and diseases), cats have the potential to be a spontaneous model for Alzheimer's disease. Classic animal models in many cases fail to predict the results in humans, and a natural model can lead to better prediction of results, thus being both time and cost-effective. The feline disease can be researched in trials that could be simultaneously clinical trials for cats and preclinical trials for humans, also referred to as reverse translational medicine. As both maladies lack effective medical intervention, new potential treatments are merited. Cannabidiol (CBD) is a promising agent that may improve the life of these patients, as it was shown to potentially treat several of the pathologies found in both conditions. yet there is a need for further research in order to establish the benefits and safety of CBD to both human and feline patients.
Collapse
Affiliation(s)
| | | | - Vered Hermush
- Department of Geriatrics and Skilled Nursing, Laniado Medical Center, Netanya, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nethanel Asher
- The Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Avi Avital
- Behavioral Neuroscience Lab, Department of Neuroscience, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Reuven Or
- Bone Marrow Transplantation, Cancer Immunotherapy & Immunobiology Research Center, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
106
|
Perpetuini D, Chiarelli AM, Filippini C, Cardone D, Croce P, Rotunno L, Anzoletti N, Zito M, Zappasodi F, Merla A. Working Memory Decline in Alzheimer's Disease Is Detected by Complexity Analysis of Multimodal EEG-fNIRS. ENTROPY (BASEL, SWITZERLAND) 2020; 22:E1380. [PMID: 33279924 PMCID: PMC7762102 DOI: 10.3390/e22121380] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is characterized by working memory (WM) failures that can be assessed at early stages through administering clinical tests. Ecological neuroimaging, such as Electroencephalography (EEG) and functional Near Infrared Spectroscopy (fNIRS), may be employed during these tests to support AD early diagnosis within clinical settings. Multimodal EEG-fNIRS could measure brain activity along with neurovascular coupling (NC) and detect their modifications associated with AD. Data analysis procedures based on signal complexity are suitable to estimate electrical and hemodynamic brain activity or their mutual information (NC) during non-structured experimental paradigms. In this study, sample entropy of whole-head EEG and frontal/prefrontal cortex fNIRS was evaluated to assess brain activity in early AD and healthy controls (HC) during WM tasks (i.e., Rey-Osterrieth complex figure and Raven's progressive matrices). Moreover, conditional entropy between EEG and fNIRS was evaluated as indicative of NC. The findings demonstrated the capability of complexity analysis of multimodal EEG-fNIRS to detect WM decline in AD. Furthermore, a multivariate data-driven analysis, performed on these entropy metrics and based on the General Linear Model, allowed classifying AD and HC with an AUC up to 0.88. EEG-fNIRS may represent a powerful tool for the clinical evaluation of WM decline in early AD.
Collapse
Affiliation(s)
- David Perpetuini
- Institute for Advanced Biomedical Technologies, Department of Neuroscience and Imaging, University G. D’Annunzio of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (A.M.C.); (C.F.); (D.C.); (P.C.); (F.Z.); (A.M.)
| | - Antonio Maria Chiarelli
- Institute for Advanced Biomedical Technologies, Department of Neuroscience and Imaging, University G. D’Annunzio of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (A.M.C.); (C.F.); (D.C.); (P.C.); (F.Z.); (A.M.)
| | - Chiara Filippini
- Institute for Advanced Biomedical Technologies, Department of Neuroscience and Imaging, University G. D’Annunzio of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (A.M.C.); (C.F.); (D.C.); (P.C.); (F.Z.); (A.M.)
| | - Daniela Cardone
- Institute for Advanced Biomedical Technologies, Department of Neuroscience and Imaging, University G. D’Annunzio of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (A.M.C.); (C.F.); (D.C.); (P.C.); (F.Z.); (A.M.)
| | - Pierpaolo Croce
- Institute for Advanced Biomedical Technologies, Department of Neuroscience and Imaging, University G. D’Annunzio of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (A.M.C.); (C.F.); (D.C.); (P.C.); (F.Z.); (A.M.)
| | - Ludovica Rotunno
- Department of Medicine and Science of Ageing, University G. D’Annunzio, Via Dei Vestini 31, 66100 Chieti, Italy; (L.R.); (N.A.); (M.Z.)
| | - Nelson Anzoletti
- Department of Medicine and Science of Ageing, University G. D’Annunzio, Via Dei Vestini 31, 66100 Chieti, Italy; (L.R.); (N.A.); (M.Z.)
| | - Michele Zito
- Department of Medicine and Science of Ageing, University G. D’Annunzio, Via Dei Vestini 31, 66100 Chieti, Italy; (L.R.); (N.A.); (M.Z.)
| | - Filippo Zappasodi
- Institute for Advanced Biomedical Technologies, Department of Neuroscience and Imaging, University G. D’Annunzio of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (A.M.C.); (C.F.); (D.C.); (P.C.); (F.Z.); (A.M.)
| | - Arcangelo Merla
- Institute for Advanced Biomedical Technologies, Department of Neuroscience and Imaging, University G. D’Annunzio of Chieti-Pescara, Via Luigi Polacchi 13, 66100 Chieti, Italy; (A.M.C.); (C.F.); (D.C.); (P.C.); (F.Z.); (A.M.)
| |
Collapse
|
107
|
Iannucci J, Rao HV, Grammas P. High Glucose and Hypoxia-Mediated Damage to Human Brain Microvessel Endothelial Cells Induces an Altered, Pro-Inflammatory Phenotype in BV-2 Microglia In Vitro. Cell Mol Neurobiol 2020; 42:985-996. [PMID: 33136275 PMCID: PMC8942976 DOI: 10.1007/s10571-020-00987-z] [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: 08/19/2020] [Accepted: 10/22/2020] [Indexed: 01/13/2023]
Abstract
Diabetes is strongly linked to the development of Alzheimer’s disease (AD), though the mechanisms for this enhanced risk are unclear. Because vascular inflammation is a consistent feature of both diabetes and AD, the cerebral microcirculation could be a key target for the effects of diabetes in the brain. The goal of this study is to explore whether brain endothelial cells, injured by diabetes-related insults, glucose and hypoxia, can affect inflammatory and activation processes in microglia in vitro. Human brain microvascular endothelial cells (HBMVECs) were either treated with 5 mM glucose (control), 30 mM glucose (high glucose), exposed to hypoxia, or exposed to hypoxia plus high glucose. HBMVEC-conditioned medium was then used to treat BV-2 microglia. Alterations in microglia phenotype were assessed through measurement of nitric oxide (NO), cytokine production, microglial activation state markers, and microglial phagocytosis. HBMVECs were injured by exposure to glucose and/or hypoxia, as assessed by release of LDH, interleukin (IL)-1β, and reactive oxygen species (ROS). HBMVECs injured by glucose and hypoxia induced increases in microglial production of NO, tumor necrosis factor-α (TNFα) and matrix metalloproteinase (MMP)-9. Injured HBMVECs significantly increased microglial expression of CD11c and CLEC7A, and decreased expression of the homeostatic marker P2RY12. Finally, bead uptake by BV-2 cells, an index of phagocytic ability, was elevated by conditioned media from injured HBMVECs. The demonstration that injury to brain endothelial cells by diabetic-associated insults, glucose and hypoxia, promotes microglial inflammation supports the idea that the cerebral microcirculation is a critical locus for the deleterious effects of diabetes in the AD brain.
Collapse
Affiliation(s)
- Jaclyn Iannucci
- The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, United States. .,Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA.
| | - Haripriya Vittal Rao
- The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, United States.,Wake Forest Baptist Medical Center, Winston-Salem, Wake Forest, NC, 27101, USA
| | - Paula Grammas
- The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, United States.,Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| |
Collapse
|
108
|
Visconte C, Canino J, Vismara M, Guidetti GF, Raimondi S, Pula G, Torti M, Canobbio I. Fibrillar amyloid peptides promote platelet aggregation through the coordinated action of ITAM- and ROS-dependent pathways. J Thromb Haemost 2020; 18:3029-3042. [PMID: 32790050 DOI: 10.1111/jth.15055] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/22/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Amyloid peptides Aβ40 and Aβ42, whose deposition in brain correlates with Alzheimer disease, are also present in platelets and have prothrombotic activities. OBJECTIVE In this study, we analyze the ability of Aβ peptides to form fibrils and to induce platelet activation and aggregation. METHODS Aβ40, Aβ42, and their scrambled peptides were diluted in phosphate buffered saline and fibrillogenesis was investigated by ThioflavinT and Congo Red. Aggregation, protein phosphorylation, and reactive oxygen species (ROS) production were analyzed. RESULTS Aβ40 and Aβ42, but not scrambled peptides, were able to form fibrils when diluted in phosphate buffered saline. Fibrillogenesis of Aβ42 was very rapid, whereas fibril formation by Aβ40 was completed only after 48 hours of incubation. Fibrillar Aβ40 and Aβ42 promoted dose-dependent aggregation of washed platelets in the presence of extracellular CaCl2 . Cleavage of GPIbα by mocarhagin or blockade of the ITAM-containing FcγRIIA prevented platelet aggregation induced by fibrillary Aβ40 and Aβ42. Fibrillar Aβ peptides stimulated the phosphorylation of FcγRIIA, resulting in the downstream stimulation of PLC, protein kinase C, and phosphoinositide 3-kinases, whose activity was necessary for full aggregation of platelets. Fibrillar Aβ peptides also induced ROS generation, and NOX inhibitors, as well as ROS scavengers, prevented platelet aggregation. However, Aβ peptide-induced ROS production did not require binding to GPIbα or activation of FcγRIIA, but was initiated by CD36, which provided an important contribution to full platelet aggregation. CONCLUSION These results suggest that fibrillar amyloid Aβ40 and Aβ42 induce platelet aggregation through the recruitment of GPIb-IX-V and CD36, which requires the convergence of ITAM- and ROS-dependent pathways.
Collapse
Affiliation(s)
- Caterina Visconte
- Neurodegenerative Disease Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Jessica Canino
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
- Scuola Universitaria Superiore, IUSS, Pavia, Italy
| | - Mauro Vismara
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | | | - Sara Raimondi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Giordano Pula
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Eppendorf (UKE), Hamburg, Germany
| | - Mauro Torti
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| |
Collapse
|
109
|
Nuthikattu S, Milenkovic D, Rutledge JC, Villablanca AC. Sex-Dependent Molecular Mechanisms of Lipotoxic Injury in Brain Microvasculature: Implications for Dementia. Int J Mol Sci 2020; 21:E8146. [PMID: 33142695 PMCID: PMC7663125 DOI: 10.3390/ijms21218146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/16/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular risk factors and biologic sex play a role in vascular dementia which is characterized by progressive reduction in cognitive function and memory. Yet, we lack understanding about the role sex plays in the molecular mechanisms whereby lipid stress contributes to cognitive decline. Five-week-old low-density lipoprotein deficient (LDL-R -/-) male and female mice and C57BL/6J wild types (WT) were fed a control or Western Diet for 8 weeks. Differential expression of protein coding and non-protein coding genes (DEG) were determined in laser captured hippocampal microvessels using genome-wide microarray, followed by bioinformatic analysis of gene networks, pathways, transcription factors and sex/gender-based analysis (SGBA). Cognitive function was assessed by Y-maze. Bioinformatic analysis revealed more DEGs in females (2412) compared to males (1972). Hierarchical clusters revealed distinctly different sex-specific gene expression profiles irrespective of diet and genotype. There were also fewer and different biologic responses in males compared to females, as well as different cellular pathways and gene networks (favoring greater neuroprotection in females), together with sex-specific transcription factors and non-protein coding RNAs. Hyperlipidemic stress also resulted in less severe cognitive dysfunction in females. This sex-specific pattern of differential hippocampal microvascular RNA expression might provide therapeutic targets for dementia in males and females.
Collapse
Affiliation(s)
- Saivageethi Nuthikattu
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| | - Dragan Milenkovic
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
- Université Clermont Auvergne, INRA, UNH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - John C. Rutledge
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| | - Amparo C. Villablanca
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| |
Collapse
|
110
|
McLarnon JG. Consideration of a Pharmacological Combinatorial Approach to Inhibit Chronic Inflammation in Alzheimer's Disease. Curr Alzheimer Res 2020; 16:1007-1017. [PMID: 31692444 DOI: 10.2174/1567205016666191106095038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/26/2019] [Accepted: 10/18/2019] [Indexed: 01/14/2023]
Abstract
A combinatorial cocktail approach is suggested as a rationale intervention to attenuate chronic inflammation and confer neuroprotection in Alzheimer's disease (AD). The requirement for an assemblage of pharmacological compounds follows from the host of pro-inflammatory pathways and mechanisms present in activated microglia in the disease process. This article suggests a starting point using four compounds which present some differential in anti-inflammatory targets and actions but a commonality in showing a finite permeability through Blood-brain Barrier (BBB). A basis for firstchoice compounds demonstrated neuroprotection in animal models (thalidomide and minocycline), clinical trial data showing some slowing in the progression of pathology in AD brain (ibuprofen) and indirect evidence for putative efficacy in blocking oxidative damage and chemotactic response mediated by activated microglia (dapsone). It is emphasized that a number of candidate compounds, other than ones suggested here, could be considered as components of the cocktail approach and would be expected to be examined in subsequent work. In this case, systematic testing in AD animal models is required to rigorously examine the efficacy of first-choice compounds and replace ones showing weaker effects. This protocol represents a practical approach to optimize the reduction of microglial-mediated chronic inflammation in AD pathology. Subsequent work would incorporate the anti-inflammatory cocktail delivery as an adjunctive treatment with ones independent of inflammation as an overall preventive strategy to slow the progression of AD.
Collapse
Affiliation(s)
- James G McLarnon
- Department of Anesthesiology, Pharmacology and Therapeutics, The University of British Columbia, Vancouver, British Columbia, V6T1Z3, Canada
| |
Collapse
|
111
|
Di Costanzo A, Paris D, Melck D, Angiolillo A, Corso G, Maniscalco M, Motta A. Blood biomarkers indicate that the preclinical stages of Alzheimer's disease present overlapping molecular features. Sci Rep 2020; 10:15612. [PMID: 32973179 PMCID: PMC7515866 DOI: 10.1038/s41598-020-71832-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
It is still debated whether non-specific preclinical symptoms of Alzheimer's disease (AD) can have diagnostic relevance. We followed the evolution from cognitively normal to AD by NMR-based metabolomics of blood sera. Multivariate statistical analysis of the NMR profiles yielded models that discriminated subjective memory decline (SMD), mild cognitive impairment (MCI) and AD. We validated a panel of six statistically significant metabolites that predicted SMD, MCI and AD in a blind cohort with sensitivity values ranging from 88 to 95% and receiver operating characteristic values from 0.88 to 0.99. However, lower values of specificity, accuracy and precision were observed for the models involving SMD and MCI, which is in line with the pathological heterogeneity indicated by clinical data. This excludes a "linear" molecular evolution of the pathology, pointing to the presence of overlapping "gray-zones" due to the reciprocal interference of the intermediate stages. Yet, the clear difference observed in the metabolic pathways of each model suggests that pathway dysregulations could be investigated for diagnostic purposes.
Collapse
Affiliation(s)
- Alfonso Di Costanzo
- 1Centre for Research and Training in Medicine for Aging, Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100, Campobasso, Italy
| | - Debora Paris
- Institute of Biomolecular Chemistry, National Research Council, 80078, Pozzuoli, Naples, Italy.
| | - Dominique Melck
- Institute of Biomolecular Chemistry, National Research Council, 80078, Pozzuoli, Naples, Italy
| | - Antonella Angiolillo
- 1Centre for Research and Training in Medicine for Aging, Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100, Campobasso, Italy
| | - Gaetano Corso
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggia, Italy
| | - Mauro Maniscalco
- Pulmonary Rehabilitation Unit, ICS Maugeri SpA SB, Institute of Telese Terme, 82037, Telese Terme, Benevento, Italy
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, 80078, Pozzuoli, Naples, Italy.
| |
Collapse
|
112
|
Zhao A, Li Y, Deng Y. TNF receptors are associated with tau pathology and conversion to Alzheimer's dementia in subjects with mild cognitive impairment. Neurosci Lett 2020; 738:135392. [PMID: 32947003 DOI: 10.1016/j.neulet.2020.135392] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/23/2020] [Accepted: 09/13/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Tumor necrosis factor-a (TNF-α) signaling pathway plays a significant role in Alzheimer's disease (AD). This study aimed to explore the relationship between TNF-α related inflammatory proteins and pathological markers of AD, and examine their possibility as a predictor of the conversion of mild cognitive impairment (MCI) to AD. METHODS This study included both cross-sectional and longitudinal designs. The levels of TNF-α related inflammatory proteins, Aβ1-42, total-tau(t-tau), phosphorylated tau (p-tau) from cerebrospinal fluid (CSF) were analyzed in healthy controls (HC, n = 90), MCI (n = 116), and AD participants (n = 75) from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Kaplan-Meier analyses were used to evaluate the predictive value of the examined putative AD markers after follow-up visits. RESULTS In the cross-sectional cohort, we observed higher CSF levels of TNF-α related inflammatory proteins in the MCI and AD patients with positive tau pathology. TNF receptors (TNFR) were more closely associated with t-tau and p-tau than Aβ1-42, in HC, MCI and AD subjects. In the longitudinal cohort with a mean follow-up of 30.2 months, MCI patients with high levels of CSF TNFR1 (p = 0.001) and low levels of TNFR2 (p < 0.001) were more likely to develop into AD. CONCLUSION TNFR-signaling might be involved in the early pathogenesis of AD and TNF receptors may serve as potential predictive biomarkers for MCI.
Collapse
Affiliation(s)
- Aonan Zhao
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuanyuan Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yulei Deng
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Department of Neurology, RuiJin Hospital/Lu Wan Branch, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China.
| | | |
Collapse
|
113
|
Giorgi FS, Galgani A, Puglisi-Allegra S, Limanaqi F, Busceti CL, Fornai F. Locus Coeruleus and neurovascular unit: From its role in physiology to its potential role in Alzheimer's disease pathogenesis. J Neurosci Res 2020; 98:2406-2434. [PMID: 32875628 DOI: 10.1002/jnr.24718] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/26/2020] [Accepted: 08/08/2020] [Indexed: 12/15/2022]
Abstract
Locus coeruleus (LC) is the main noradrenergic (NA) nucleus of the central nervous system. LC degenerates early during Alzheimer's disease (AD) and NA loss might concur to AD pathogenesis. Aside from neurons, LC terminals provide dense innervation of brain intraparenchymal arterioles/capillaries, and NA modulates astrocyte functions. The term neurovascular unit (NVU) defines the strict anatomical/functional interaction occurring between neurons, glial cells, and brain vessels. NVU plays a fundamental role in coupling the energy demand of activated brain regions with regional cerebral blood flow, it includes the blood-brain barrier (BBB), plays an active role in neuroinflammation, and participates also to the glymphatic system. NVU alteration is involved in AD pathophysiology through several mechanisms, mainly related to a relative oligoemia in activated brain regions and impairment of structural and functional BBB integrity, which contributes also to the intracerebral accumulation of insoluble amyloid. We review the existing data on the morphological features of LC-NA innervation of the NVU, as well as its contribution to neurovascular coupling and BBB proper functioning. After introducing the main experimental data linking LC with AD, which have repeatedly shown a key role of neuroinflammation and increased amyloid plaque formation, we discuss the potential mechanisms by which the loss of NVU modulation by LC might contribute to AD pathogenesis. Surprisingly, thus far not so many studies have tested directly these mechanisms in models of AD in which LC has been lesioned experimentally. Clarifying the interaction of LC with NVU in AD pathogenesis may disclose potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Filippo Sean Giorgi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Neurology Unit, Pisa University Hospital, Pisa, Italy
| | | | | | - Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,I.R.C.C.S. I.N.M. Neuromed, Pozzilli, Italy
| |
Collapse
|
114
|
Migrino RQ, Karamanova N, Truran S, Serrano GE, Davies HA, Madine J, Beach TG. Cerebrovascular medin is associated with Alzheimer's disease and vascular dementia. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12072. [PMID: 32875054 PMCID: PMC7447901 DOI: 10.1002/dad2.12072] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/18/2020] [Accepted: 06/24/2020] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Medin, an aging-associated amyloidogenic protein, induces cerebrovascular dysfunction and inflammation. We investigated the relationship between cerebrovascular medin and Alzheimer's disease (AD) and vascular dementia (VaD). METHODS Cerebral arteriole medin was quantified from 91 brain donors with no dementia (ND), AD, VaD, or combined AD and VaD. Correlation analyses evaluated the relationship between arteriole medin, and plaques, tangles, or white matter lesions (WML). Receiver operating characteristic and regression analyses assessed whether medin is predictive of AD or VaD versus other cerebrovascular pathologies (circle of Willis [CoW] atherosclerosis and cerebral amyloid angiopathy [CAA]). RESULTS Arteriole medin was higher in those with AD, VaD, or combined AD/VaD versus ND (P < .05), and correlated with tangle, plaque, and WML, but not CAA or CoW atherosclerosis. Among cerebrovascular pathologies, medin was the strongest predictor of AD diagnosis, whereas CoW atherosclerosis and arteriole medin were predictors of VaD. DISCUSSION Cerebral arteriole medin is associated with and could be a potential novel risk factor or biomarker for AD and VaD.
Collapse
Affiliation(s)
- Raymond Q. Migrino
- Phoenix Veterans Affairs Health Care SystemPhoenixArizonaUSA
- University of Arizona College of Medicine‐PhoenixPhoenixArizonaUSA
| | - Nina Karamanova
- Phoenix Veterans Affairs Health Care SystemPhoenixArizonaUSA
| | - Seth Truran
- Phoenix Veterans Affairs Health Care SystemPhoenixArizonaUSA
| | | | | | | | | |
Collapse
|
115
|
Bennett RE, Hu M, Fernandes A, Perez-Rando M, Robbins A, Kamath T, Dujardin S, Hyman BT. Tau reduction in aged mice does not impact Microangiopathy. Acta Neuropathol Commun 2020; 8:137. [PMID: 32811565 PMCID: PMC7436970 DOI: 10.1186/s40478-020-01014-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 11/30/2022] Open
Abstract
Microangiopathy, including proliferation of small diameter capillaries, increasing vessel tortuosity, and increased capillary blockage by leukocytes, was previously observed in the aged rTg4510 mouse model. Similar gene expression changes related to angiogenesis were observed in both rTg4510 and Alzheimer's disease (AD). It is uncertain if tau is directly responsible for these vascular changes by interacting directly with microvessels, and/or if it contributes indirectly via neurodegeneration and concurrent neuronal loss and inflammation. To better understand the nature of tau-related microangiopathy in human AD and in tau mice, we isolated capillaries and observed that bioactive soluble tau protein could be readily detected in association with vasculature. To examine whether this soluble tau is directly responsible for the microangiopathic changes, we made use of the tetracycline-repressible gene expression cassette in the rTg4510 mouse model and measured vascular pathology following tau reduction. These data suggest that reduction of tau is insufficient to alter established microvascular complications including morphological alterations, enhanced expression of inflammatory genes involved in leukocyte adherence, and blood brain barrier compromise. These data imply that 1) soluble bioactive tau surprisingly accumulates at the blood brain barrier in human brain and in mouse models, and 2) the morphological and molecular phenotype of microvascular disturbance does not resolve with reduction of whole brain soluble tau. Additional consideration of vascular-directed therapies and strategies that target tau in the vascular space may be required to restore normal function in neurodegenerative disease.
Collapse
Affiliation(s)
- Rachel E Bennett
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Miwei Hu
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Analiese Fernandes
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Marta Perez-Rando
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ashley Robbins
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Tarun Kamath
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Simon Dujardin
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Bradley T Hyman
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| |
Collapse
|
116
|
Iannucci J, Renehan W, Grammas P. Thrombin, a Mediator of Coagulation, Inflammation, and Neurotoxicity at the Neurovascular Interface: Implications for Alzheimer's Disease. Front Neurosci 2020; 14:762. [PMID: 32792902 PMCID: PMC7393221 DOI: 10.3389/fnins.2020.00762] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022] Open
Abstract
The societal burden of Alzheimer’s disease (AD) is staggering, with current estimates suggesting that 50 million people world-wide have AD. Identification of new therapeutic targets is a critical barrier to the development of disease-modifying therapies. A large body of data implicates vascular pathology and cardiovascular risk factors in the development of AD, indicating that there are likely shared pathological mediators. Inflammation plays a role in both cardiovascular disease and AD, and recent evidence has implicated elements of the coagulation system in the regulation of inflammation. In particular, the multifunctional serine protease thrombin has been found to act as a mediator of vascular dysfunction and inflammation in both the periphery and the central nervous system. In the periphery, thrombin contributes to the development of cardiovascular disease, including atherosclerosis and diabetes, by inducing endothelial dysfunction and related inflammation. In the brain, thrombin has been found to act on endothelial cells of the blood brain barrier, microglia, astrocytes, and neurons in a manner that promotes vascular dysfunction, inflammation, and neurodegeneration. Thrombin is elevated in the AD brain, and thrombin signaling has been linked to both tau and amyloid beta, pathological hallmarks of the disease. In AD mouse models, inhibiting thrombin preserves cognition and endothelial function and reduces neuroinflammation. Evidence linking atrial fibrillation with AD and dementia indicates that anticoagulant therapy may reduce the risk of dementia, with targeting thrombin shown to be particularly effective. It is time for “outside-the-box” thinking about how vascular risk factors, such as atherosclerosis and diabetes, as well as the coagulation and inflammatory pathways interact to promote increased AD risk. In this review, we present evidence that thrombin is a convergence point for AD risk factors and as such that thrombin-based therapeutics could target multiple points of AD pathology, including neurodegeneration, vascular activation, and neuroinflammation. The urgent need for disease-modifying drugs in AD demands new thinking about disease pathogenesis and an exploration of novel drug targets, we propose that thrombin inhibition is an innovative tactic in the therapeutic battle against this devastating disease.
Collapse
Affiliation(s)
- Jaclyn Iannucci
- The George and Anne Ryan Institute for Neuroscience, The University of Rhode Island, Kingston, RI, United States.,Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, RI, United States
| | - William Renehan
- The George and Anne Ryan Institute for Neuroscience, The University of Rhode Island, Kingston, RI, United States
| | - Paula Grammas
- The George and Anne Ryan Institute for Neuroscience, The University of Rhode Island, Kingston, RI, United States.,Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, The University of Rhode Island, Kingston, RI, United States
| |
Collapse
|
117
|
Cervellati C, Trentini A, Pecorelli A, Valacchi G. Inflammation in Neurological Disorders: The Thin Boundary Between Brain and Periphery. Antioxid Redox Signal 2020; 33:191-210. [PMID: 32143546 DOI: 10.1089/ars.2020.8076] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Accumulating evidence suggests that inflammation is a major contributor in the pathogenesis of several highly prevalent, but also rare, neurological diseases. In particular, the neurodegenerative processes of Alzheimer's disease (AD), vascular dementia (VAD), Parkinson's disease (PD), and multiple sclerosis (MS) are fueled by neuroinflammation, which, in turn, is accompanied by a parallel systemic immune dysregulation. This cross-talk between periphery and the brain becomes substantial when the blood-brain barrier loses its integrity, as often occurs in the course of these diseases. It has been hypothesized that the perpetual bidirectional flux of inflammatory mediators is not a mere "static" collateral effect of the neurodegeneration, but represents a proactive phenomenon sparking and driving the neuropathological processes. However, the upstream/downstream relationship between inflammatory events and neurological pathology is still unclear. Recent Advances: Solid recent evidence clearly suggests that metabolic factors, systemic infections, Microbiota dysbiosis, and oxidative stress are implicated, although to a different extent, in the development in brain diseases. Critical Issues: Here, we reviewed the most solid published evidence supporting the implication of the axis systemic inflammation-neuroinflammation-neurodegeneration in the pathogenesis of AD, VAD, PD, and MS, highlighting the possible cause of the putative downstream component of the axis. Future Directions: Reaching a definitive clinical/epidemiological appreciation of the etiopathogenic significance of the connection between peripheral and brain inflammation in neurologic disorders is pivotal since it could open novel therapeutic avenues for these diseases.
Collapse
Affiliation(s)
- Carlo Cervellati
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandro Trentini
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandra Pecorelli
- Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina, USA
| | - Giuseppe Valacchi
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy.,Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina, USA.,Department of Food and Nutrition, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
118
|
Miller LM, Jenny NS, Rawlings AM, Arnold AM, Fitzpatrick AL, Lopez OL, Odden MC. Sex Differences in the Association Between Pentraxin 3 and Cognitive Decline: The Cardiovascular Health Study. J Gerontol A Biol Sci Med Sci 2020; 75:1523-1529. [PMID: 31808814 PMCID: PMC7357589 DOI: 10.1093/gerona/glz217] [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: 03/07/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The importance of systemic inflammation, measured by C-reactive protein, in cognitive decline has been demonstrated; however, the role of vascular inflammation is less understood. Pentraxin 3 (PTX3) is a novel marker of vascular inflammation. METHODS We followed adults 65 and older, free of cardiovascular disease (CVD) for up to 9 years (n = 1,547) in the Cardiovascular Health Study. We evaluated the relationship between PTX3 and change in cognitive function, measured using the Modified Mini-Mental State Examination (3MSE), and incident cognitive impairment (3MSE < 80). Mediation by CVD events, and effect modification by sex and apolipoprotein E ɛ4 allele (APOE4) were also examined. RESULTS The average decline in 3MSE was 0.77 points per year. The association between PTX3 and change in 3MSE differed between women and men (p = .02). In the adjusted model, each standard deviation higher in PTX3 was associated with a 0.20 greater decline in 3MSE score per year in women over follow-up (95% CI: -0. 37, -0.03; p = .02), compared to no change in men (β = 0.07; 95% CI: -0.08, 0.22). CVD events had a minor effect on the associations. No effect modification by APOE4 was found, although we observed the association of PTX3 and cognitive impairment in women was attenuated and nonsignificant after adjustment for APOE4. There was a paradoxical protective association between PTX3 and reduced cognitive impairment in men, even after adjustment for APOE4. CONCLUSIONS We found that vascular inflammation was significantly associated with cognitive decline in older women, but not men.
Collapse
Affiliation(s)
- Lindsay M Miller
- Division of Nephrology-Hypertension, Department of Medicine, University of San Diego, La Jolla
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis
| | - Nancy S Jenny
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington
| | - Andreea M Rawlings
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis
- Kaiser Permanente Center for Health Research, Portland, Oregon
| | - Alice M Arnold
- Department of Biostatistics, University of Washington, Seattle
| | - Annette L Fitzpatrick
- Departments of Family Medicine, Epidemiology, and Global Health, University of Washington, Seattle
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pennsylvania
| | - Michelle C Odden
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis
- Department of Health Research and Policy, Stanford University, Stanford, California
| |
Collapse
|
119
|
Mészáros Á, Molnár K, Nógrádi B, Hernádi Z, Nyúl-Tóth Á, Wilhelm I, Krizbai IA. Neurovascular Inflammaging in Health and Disease. Cells 2020; 9:cells9071614. [PMID: 32635451 PMCID: PMC7407516 DOI: 10.3390/cells9071614] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/02/2020] [Indexed: 12/19/2022] Open
Abstract
Aging is characterized by a chronic low-grade sterile inflammation dubbed as inflammaging, which in part originates from accumulating cellular debris. These, acting as danger signals with many intrinsic factors such as cytokines, are sensed by a network of pattern recognition receptors and other cognate receptors, leading to the activation of inflammasomes. Due to the inflammasome activity-dependent increase in the levels of pro-inflammatory interleukins (IL-1β, IL-18), inflammation is initiated, resulting in tissue injury in various organs, the brain and the spinal cord included. Similarly, in age-related diseases of the central nervous system (CNS), inflammasome activation is a prominent moment, in which cells of the neurovascular unit occupy a significant position. In this review, we discuss the inflammatory changes in normal aging and summarize the current knowledge on the role of inflammasomes and contributing mechanisms in common CNS diseases, namely Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis and stroke, all of which occur more frequently with aging.
Collapse
Affiliation(s)
- Ádám Mészáros
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Doctoral School of Biology, University of Szeged, 6726 Szeged, Hungary
| | - Kinga Molnár
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Theoretical Medicine Doctoral School, University of Szeged, 6720 Szeged, Hungary
| | - Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, 6720 Szeged, Hungary
| | - Zsófia Hernádi
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, 6720 Szeged, Hungary
| | - Ádám Nyúl-Tóth
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania
| | - István A. Krizbai
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania
- Correspondence: ; Tel.: +36-62-599-794
| |
Collapse
|
120
|
Shabani S, Rabiei Z, Amini-Khoei H. Exploring the multifaceted neuroprotective actions of gallic acid: a review. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2020. [DOI: 10.1080/10942912.2020.1753769] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sahreh Shabani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Rabiei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
121
|
Hindam MO, Sayed RH, Skalicka-Woźniak K, Budzyńska B, El Sayed NS. Xanthotoxin and umbelliferone attenuate cognitive dysfunction in a streptozotocin-induced rat model of sporadic Alzheimer's disease: The role of JAK2/STAT3 and Nrf2/HO-1 signalling pathway modulation. Phytother Res 2020; 34:2351-2365. [PMID: 32250498 DOI: 10.1002/ptr.6686] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/05/2020] [Accepted: 03/14/2020] [Indexed: 12/18/2022]
Abstract
The aim of the present study was to assess the neuroprotective effects of xanthotoxin and umbelliferone in streptozotocin (STZ)-induced cognitive dysfunction in rats. Animals were injected intracerebroventricularly (ICV) with STZ (3 mg/kg) once to induce a sporadic Alzheimer's disease (SAD)-like condition. Xanthotoxin or umbelliferone (15 mg/kg, i.p.) were administered 5 hr after ICV-STZ and daily for 20 consecutive days. Xanthotoxin or umbelliferone prevented cognitive deficits in the Morris water maze and object recognition tests. In parallel, xanthotoxin or umbelliferone reduced hippocampal acetylcholinestrase activity and malondialdehyde level. Moreover, xanthotoxin or umbelliferone increased glutathione content. These coumarins also modulated neuronal cell death by reducing the level of proinflammatory cytokines (tumour necrosis factor-alpha and interleukin-6), inhibiting the overexpression of inflammatory markers (nuclear factor κB [NF-κB] and cyclooxygenase II), and upregulating the expression of NF-κB inhibitor (IκB-α). Interestingly, xanthotoxin diminished phosphorylated JAK2 and phosphorylated STAT3 protein expression, while umbelliferone markedly replenished nuclear factor erythroid-derived 2-like 2 (Nrf2) and haem oxygenase-1 (HO-1) levels. The current study provides evidence for the protective effect of xanthotoxin and umbelliferone in STZ-induced cognitive dysfunction in rats. This effect may be attributed, at least in part, to inhibiting acetylcholinestrase and attenuating oxidative stress, neuroinflammation and neuronal loss.
Collapse
Affiliation(s)
- Merhan O Hindam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | | | - Barbara Budzyńska
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, Lublin, Poland
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
122
|
Smith PJ. Pathways of Prevention: A Scoping Review of Dietary and Exercise Interventions for Neurocognition. Brain Plast 2019; 5:3-38. [PMID: 31970058 PMCID: PMC6971820 DOI: 10.3233/bpl-190083] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease and related dementias (ADRD) represent an increasingly urgent public health concern, with an increasing number of baby boomers now at risk. Due to a lack of efficacious therapies among symptomatic older adults, an increasing emphasis has been placed on preventive measures that can curb or even prevent ADRD development among middle-aged adults. Lifestyle modification using aerobic exercise and dietary modification represents one of the primary treatment modalities used to mitigate ADRD risk, with an increasing number of trials demonstrating that exercise and dietary change, individually and together, improve neurocognitive performance among middle-aged and older adults. Despite several optimistic findings, examination of treatment changes across lifestyle interventions reveals a variable pattern of improvements, with large individual differences across trials. The present review attempts to synthesize available literature linking lifestyle modification to neurocognitive changes, outline putative mechanisms of treatment improvement, and discuss discrepant trial findings. In addition, previous mechanistic assumptions linking lifestyle to neurocognition are discussed, with a focus on potential solutions to improve our understanding of individual neurocognitive differences in response to lifestyle modification. Specific recommendations include integration of contemporary causal inference approaches for analyzing parallel mechanistic pathways and treatment-exposure interactions. Methodological recommendations include trial multiphase optimization strategy (MOST) design approaches that leverage individual differences for improved treatment outcomes.
Collapse
Affiliation(s)
- Patrick J. Smith
- Department of Psychiatry and Behavioral Sciences (Primary), Duke University Medical Center, NC, USA
- Department of Medicine (Secondary), Duke University Medical Center, NC, USA
- Department of Population Health Sciences (Secondary), Duke University, NC, USA
| |
Collapse
|
123
|
Voluntary exercise increases brain tissue oxygenation and spatially homogenizes oxygen delivery in a mouse model of Alzheimer's disease. Neurobiol Aging 2019; 88:11-23. [PMID: 31866158 DOI: 10.1016/j.neurobiolaging.2019.11.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 11/21/2022]
Abstract
Although vascular contributions to dementia and Alzheimer's disease (AD) are increasingly recognized, the potential brain oxygenation disruption associated with AD and whether preventive strategies to maintain tissue oxygenation are beneficial remain largely unknown. This study aimed to examine (1) whether brain oxygenation is compromised by the onset of AD and (2) how voluntary exercise modulates the influence of AD on brain oxygenation. In vivo 2-photon phosphorescence lifetime microscopy was used to investigate local changes of brain tissue oxygenation with the progression of AD and its modulation by exercise in the barrel cortex of awake transgenic AD mice. Our results show that cerebral tissue oxygen partial pressure (PO2) decreased with the onset of AD. Reduced PO2 was associated with the presence of small near-hypoxic areas, an increased oxygen extraction fraction, and reduced blood flow, observations that were all reverted by exercise. AD and age also increased the spatial heterogeneity of brain tissue oxygenation, which was normalized by exercise. Ex vivo staining also showed fewer amyloid-β (Aβ) deposits in the exercise group. Finally, we observed correlations between voluntary running distance and cerebral tissue oxygenation/blood flow, suggesting a dose-response relationship of exercise on the brain. Overall, this study suggests that compromised brain oxygenation is an indicator of the onset of AD, with the emergence of potential deleterious mechanisms associated with hypoxia. Furthermore, voluntary exercise enhanced the neurovascular oxygenation process, potentially offering a means to delay these changes.
Collapse
|
124
|
Kim CM, Alvarado RL, Stephens K, Wey HY, Wang DJJ, Leritz EC, Salat DH. Associations between cerebral blood flow and structural and functional brain imaging measures in individuals with neuropsychologically defined mild cognitive impairment. Neurobiol Aging 2019; 86:64-74. [PMID: 31813626 DOI: 10.1016/j.neurobiolaging.2019.10.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/05/2019] [Accepted: 10/08/2019] [Indexed: 01/18/2023]
Abstract
Reduced cerebral blood flow (CBF), an indicator of neurovascular processes and metabolic demands, is a common finding in Alzheimer's disease. However, little is known about what contributes to CBF deficits in individuals with mild cognitive impairment (MCI). We examine regional CBF differences in 17 MCI compared with 21 age-matched cognitively healthy older adults. Next, we examined associations between CBF, white matter lesion (WML) volume, amplitude of low-frequency fluctuations, and cortical thickness to better understand whether altered CBF was detectable before other markers and the potential mechanistic underpinnings of CBF deficits in MCI. MCI had significantly reduced CBF, whereas cortical thickness and amplitude of low-frequency fluctuation were not affected. Reduced CBF was associated with the WML volume but not associated with other measures. Given the presumed vascular etiology of WML and relative worsening of vascular health in MCI, it may suggest CBF deficits result from early vascular as opposed to metabolic deficits in MCI. These findings may support vascular mechanisms as an underlying component of cognitive impairment.
Collapse
Affiliation(s)
- Chan-Mi Kim
- Brain Aging and Dementia (BAnD) Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Rachel L Alvarado
- Brain Aging and Dementia (BAnD) Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Kimberly Stephens
- Brain Aging and Dementia (BAnD) Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Hsiao-Ying Wey
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Dany J J Wang
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, CA, USA; Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth C Leritz
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Geriatric Research, Education & Clinical Center & Translational Research Center for TBI and Stress Disorders, VA Boston Healthcare System, Boston, MA, USA
| | - David H Salat
- Brain Aging and Dementia (BAnD) Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Neuroimaging Research for Veterans (NeRVe) Center, VA Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
125
|
Nyunt T, Britton M, Wanichthanarak K, Budamagunta M, Voss JC, Wilson DW, Rutledge JC, Aung HH. Mitochondrial oxidative stress-induced transcript variants of ATF3 mediate lipotoxic brain microvascular injury. Free Radic Biol Med 2019; 143:25-46. [PMID: 31356870 PMCID: PMC6848793 DOI: 10.1016/j.freeradbiomed.2019.07.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/30/2019] [Accepted: 07/23/2019] [Indexed: 12/06/2022]
Abstract
Elevation of blood triglycerides, primarily triglyceride-rich lipoproteins (TGRL), is an independent risk factor for cardiovascular disease and vascular dementia (VaD). Accumulating evidence indicates that both atherosclerosis and VaD are linked to vascular inflammation. However, the role of TGRL in vascular inflammation, which increases risk for VaD, remains largely unknown and its underlying mechanisms are still unclear. We strived to determine the effects of postprandial TGRL exposure on brain microvascular endothelial cells, the potential risk factor of vascular inflammation, resulting in VaD. We showed in Aung et al., J Lipid Res., 2016 that postprandial TGRL lipolysis products (TL) activate mitochondrial reactive oxygen species (ROS) and increase the expression of the stress-responsive protein, activating transcription factor 3 (ATF3), which injures human brain microvascular endothelial cells (HBMECs) in vitro. In this study, we deployed high-throughput sequencing (HTS)-based RNA sequencing methods and mito stress and glycolytic rate assays with an Agilent Seahorse XF analyzer and profiled the differential expression of transcripts, constructed signaling pathways, and measured mitochondrial respiration, ATP production, proton leak, and glycolysis of HBMECs treated with TL. Conclusions: TL potentiate ROS by mitochondria which activate mitochondrial oxidative stress, decrease ATP production, increase mitochondrial proton leak and glycolysis rate, and mitochondria DNA damage. Additionally, CPT1A1 siRNA knockdown suppresses oxidative stress and prevents mitochondrial dysfunction and vascular inflammation in TL treated HBMECs. TL activates ATF3-MAPKinase, TNF, and NRF2 signaling pathways. Furthermore, the NRF2 signaling pathway which is upstream of the ATF3-MAPKinase signaling pathway, is also regulated by the mitochondrial oxidative stress. We are the first to report differential inflammatory characteristics of transcript variants 4 (ATF3-T4) and 5 (ATF3-T5) of the stress responsive gene ATF3 in HBMECs induced by postprandial TL. Specifically, our data indicates that ATF3-T4 predominantly regulates the TL-induced brain microvascular inflammation and TNF signaling. Both siRNAs of ATF3-T4 and ATF3-T5 suppress cells apoptosis and lipotoxic brain microvascular endothelial cells. These novel signaling pathways triggered by oxidative stress-responsive transcript variants, ATF3-T4 and ATF3-T5, in the brain microvascular inflammation induced by TGRL lipolysis products may contribute to pathophysiological processes of vascular dementia.
Collapse
Affiliation(s)
- Tun Nyunt
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Monica Britton
- Genome Center and Bioinformatics Core Facility, University of California, Davis, CA, 95616, USA
| | - Kwanjeera Wanichthanarak
- West Coast Metabolomics Center, Genome Center, University of California Davis, Davis, CA, USA; Department of Biochemistry and Siriraj Metabolomics and Phenomics Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Madhu Budamagunta
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - John C Voss
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Dennis W Wilson
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - John C Rutledge
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Hnin H Aung
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
126
|
Abstract
Small vessel disease (SVD) refers to conditions where damage to arterioles and capillaries is predominant, leading to reduced, or interrupted perfusion of the affected organ. Data suggest that when this condition is evident in any organ, it is already systemic in its occurrence and consequences. SVD affects primarily organs that receive significant portions of cardiac output such as the brain, the kidney, and the retina. Thus, SVD is a major etiologic cause in debilitating conditions such as renal failure, blindness, lacunar infarcts, and dementia. The factors that lead to this devastating condition include all the known vascular risk factors when they are not strictly controlled, but lifestyles that include sedentary existence, obesity, and poor sleep patterns are also recognized drivers of SVD. In addition, depression is now recognized as a vascular risk factor. Inflammation is a mediator of SVD, but it is not known which factor(s) predominate in its etiology. This article emphasizes the need for more investigations to define this link further and suggests clinical and societal responses that might reduce the major impacts of this condition on populations.
Collapse
Affiliation(s)
- Antoine M Hakim
- Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.,Division of Neurology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
127
|
Espinosa-Parrilla Y, Gonzalez-Billault C, Fuentes E, Palomo I, Alarcón M. Decoding the Role of Platelets and Related MicroRNAs in Aging and Neurodegenerative Disorders. Front Aging Neurosci 2019; 11:151. [PMID: 31312134 PMCID: PMC6614495 DOI: 10.3389/fnagi.2019.00151] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/11/2019] [Indexed: 12/22/2022] Open
Abstract
Platelets are anucleate cells that circulate in blood and are essential components of the hemostatic system. During aging, platelet numbers decrease and their aggregation capacity is reduced. Platelet dysfunctions associated with aging can be linked to molecular alterations affecting several cellular systems that include cytoskeleton rearrangements, signal transduction, vesicular trafficking, and protein degradation. Age platelets may adopt a phenotype characterized by robust secretion of extracellular vesicles that could in turn account for about 70-90% of blood circulating vesicles. Interestingly these extracellular vesicles are loaded with messenger RNAs and microRNAs that may have a profound impact on protein physiology at the systems level. Age platelet dysfunction is also associated with accumulation of reactive oxygen species. Thereby understanding the mechanisms of aging in platelets as well as their age-dependent dysfunctions may be of interest when evaluating the contribution of aging to the onset of age-dependent pathologies, such as those affecting the nervous system. In this review we summarize the findings that link platelet dysfunctions to neurodegenerative diseases including Alzheimer's Disease, Parkinson's Disease, Multiple Sclerosis, Huntington's Disease, and Amyotrophic Lateral Sclerosis. We discuss the role of platelets as drivers of protein dysfunctions observed in these pathologies, their association with aging and the potential clinical significance of platelets, and related miRNAs, as peripheral biomarkers for diagnosis and prognosis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yolanda Espinosa-Parrilla
- School of Medicine, Universidad de Magallanes, Punta Arenas, Chile
- Laboratory of Molecular Medicine-LMM, Center for Education, Healthcare and Investigation-CADI, Universidad de Magallanes, Punta Arenas, Chile
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
| | - Christian Gonzalez-Billault
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism GERO, Santiago, Chile
- The Buck Institute for Research on Aging, Novato, CA, United States
| | - Eduardo Fuentes
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences and Research Center for Aging, Universidad de Talca, Talca, Chile
| | - Ivan Palomo
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences and Research Center for Aging, Universidad de Talca, Talca, Chile
| | - Marcelo Alarcón
- Thematic Task Force on Healthy Aging, CUECH Research Network, Santiago, Chile
- Thrombosis Research Center, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences and Research Center for Aging, Universidad de Talca, Talca, Chile
| |
Collapse
|
128
|
Sung CE, Huang RY, Cheng WC, Kao TW, Chen WL. Association between periodontitis and cognitive impairment: Analysis of national health and nutrition examination survey (NHANES) III. J Clin Periodontol 2019; 46:790-798. [PMID: 31152592 DOI: 10.1111/jcpe.13155] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/21/2019] [Accepted: 05/26/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Periodontitis has been hypothesized as being one of the most common potential risk factors for the development of dementia and cognitive impairment. In order to investigate the relationship between periodontitis and cognition impairment, the National Health and Nutrition Examination Survey (NHANES) database was analysed after adjusting for potential confounding factors, including age and other systemic co-morbidities. MATERIALS AND METHODS In total, 4,663 participants aged 20-59 years who had received full-mouth periodontal examination and undergone the cognitive functional test were enrolled. The grade of periodontal disease was categorized into severe, moderate, and mild. Cognitive function examinations, including the simple reaction time test (SRTT), symbol digit substitution test (SDST), and serial digit learning test (SDLT), were adopted for the evaluation of cognitive impairment. RESULTS The subjects with mild and moderate to severe periodontitis had higher SDLT and SDST scores, which indicated decreased cognitive function, compared with the healthy group. After adjusting for demographic factors, education, smoking, cardiovascular diseases, and laboratory data, periodontitis was significantly correlated with elevated SDST and SDLT scores (p values for trend = 0.014 and 0.038, respectively) by generalized linear regression models. CONCLUSION Our study highlighted that periodontal status was associated with cognitive impairment in a nationally representative sample of US adults.
Collapse
Affiliation(s)
- Cheng-En Sung
- Department of Periodontology, School of Dentistry, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
| | - Ren-Yeong Huang
- Department of Periodontology, School of Dentistry, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
| | - Wan-Chien Cheng
- Department of Periodontology, School of Dentistry, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
| | - Tung-Wei Kao
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China.,School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Wei-Liang Chen
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan, Republic of China.,School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
129
|
Bergland AK, Soennesyn H, Dalen I, Rodriguez-Mateos A, Berge RK, Giil LM, Rajendran L, Siow R, Tassotti M, Larsen AI, Aarsland D. Effects of Anthocyanin Supplementation on Serum Lipids, Glucose, Markers of Inflammation and Cognition in Adults With Increased Risk of Dementia - A Pilot Study. Front Genet 2019; 10:536. [PMID: 31244884 PMCID: PMC6581024 DOI: 10.3389/fgene.2019.00536] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/16/2019] [Indexed: 02/04/2023] Open
Abstract
Background Anthocyanins may protect against cardiovascular related cognitive decline and dementia. Objective Open-label study to measure changes in serum lipids, glucose, glycosylated hemoglobin (HbA1c), and markers of inflammation after anthocyanin supplementation in people with increased risk of dementia. As a secondary endpoint we examined potential changes in a battery of cognitive test in the anthocyanin group (AG). A total of 27 individuals with mild cognitive impairment (MCI) (n = 8) or stable non-obstructive coronary artery disease (CAD) (n = 19) consumed two Medox® capsules, each containing 80 mg of natural purified anthocyanins, twice daily for 16 weeks. They provided blood samples and performed a short battery of cognitive tests. Twenty healthy normal controls (NC) (n = 20) provided blood samples, but did not receive any intervention and did not perform cognitive tests. Results There was a significant difference between groups for monocyte chemoattractant protein (MCP-1) and fasting glucose. In addition, total cholesterol and triglycerides were significantly increased in the AG. Improvements in memory and executive test scores were observed. No adverse effects were reported. Conclusion The results of this pilot study were largely inconclusive with regard to the potential protective effects of anthocyanin supplementation. However, anthocyanins were well tolerated, and compliance was high. Larger, placebo-controlled studies to explore the potential effects of anthocyanins on dementia risk are encouraged. Clinical Trial Registration www.ClinicalTrials.gov, identifier NCT02409446
Collapse
Affiliation(s)
- Anne Katrine Bergland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Hogne Soennesyn
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Ingvild Dalen
- Section of Biostatistics, Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London, United Kingdom
| | - Rolf Kristian Berge
- The Lipid Research Group, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lasse Melvaer Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Lawrence Rajendran
- UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Richard Siow
- School of Cardiovascular Medicine and Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Michele Tassotti
- Department of Nutritional Sciences, Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London, United Kingdom.,Department of Food & Drug, University of Parma, Parma, Italy
| | - Alf Inge Larsen
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | - Dag Aarsland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway.,UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
130
|
Huang Y, Guo B, Shi B, Gao Q, Zhou Q. Chinese Herbal Medicine Xueshuantong Enhances Cerebral Blood Flow and Improves Neural Functions in Alzheimer's Disease Mice. J Alzheimers Dis 2019; 63:1089-1107. [PMID: 29710701 PMCID: PMC6004915 DOI: 10.3233/jad-170763] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reduced cerebral blood flow in Alzheimer's disease (AD) may occur in early AD, which contributes to the pathogenesis and/or pathological progression of AD. Reversing this deficit may have therapeutic potential. Certain traditional Chinese herbal medicines (e.g., Saponin and its major component Xueshuantong [XST]) increase blood flow in humans, but whether they could be effective in treating AD patients has not been tested. We found that systemic XST injection elevated cerebral blood flow in APP/PS1 transgenic mice using two-photon time-lapse imaging in the same microvessels before and after injection. Subchronic XST treatment led to improved spatial learning and memory and motor performance in the APP/PS1 mice, suggesting improved neural plasticity and functions. Two-photon time lapse imaging of the same plaques revealed a reduction in plaque size after XST treatment. In addition, western blots experiments showed that XST treatment led to reduced processing of amyloid-β protein precursor (AβPP) and enhanced clearance of amyloid-β (Aβ) without altering the total level of AβPP. We also found increased synapse density in the immediate vicinity of amyloid plaques, suggesting enhanced synaptic function. We conclude that targeting cerebral blood flow can be an effective strategy in treating AD.
Collapse
Affiliation(s)
- Yangmei Huang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Baihong Guo
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Bihua Shi
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qingtao Gao
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| |
Collapse
|
131
|
Dong B, An L, Yang X, Zhang X, Zhang J, Tuerhong M, Jin DQ, Ohizumi Y, Lee D, Xu J, Guo Y. Withanolides from Physalis peruviana showing nitric oxide inhibitory effects and affinities with iNOS. Bioorg Chem 2019; 87:585-593. [DOI: 10.1016/j.bioorg.2019.03.051] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/22/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
|
132
|
George G, Valiya Parambath S, Lokappa SB, Varkey J. Construction of Parkinson's disease marker-based weighted protein-protein interaction network for prioritization of co-expressed genes. Gene 2019; 697:67-77. [DOI: 10.1016/j.gene.2019.02.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/12/2019] [Accepted: 02/01/2019] [Indexed: 12/31/2022]
|
133
|
Solé M, Esteban-Lopez M, Taltavull B, Fábregas C, Fadó R, Casals N, Rodríguez-Álvarez J, Miñano-Molina AJ, Unzeta M. Blood-brain barrier dysfunction underlying Alzheimer's disease is induced by an SSAO/VAP-1-dependent cerebrovascular activation with enhanced Aβ deposition. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2189-2202. [PMID: 31047972 DOI: 10.1016/j.bbadis.2019.04.016] [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] [Received: 11/20/2018] [Revised: 03/03/2019] [Accepted: 04/26/2019] [Indexed: 12/11/2022]
Abstract
Dysfunctions of the vascular system directly contribute to the onset and progression of Alzheimer's disease (AD). The blood-brain barrier (BBB) shows signs of malfunction at early stages of the disease. When Abeta peptide (Aβ) is deposited on brain vessels, it induces vascular degeneration by producing reactive oxygen species and promoting inflammation. These molecular processes are also related to an excessive SSAO/VAP-1 (semicarbazide-sensitive amine oxidase) enzymatic activity, observed in plasma and in cerebrovascular tissue of AD patients. We studied the contribution of vascular SSAO/VAP-1 to the BBB dysfunction in AD using in vitro BBB models. Our results show that SSAO/VAP-1 expression is associated to endothelial activation by altering the release of pro-inflammatory and pro-angiogenic angioneurins, most highly IL-6, IL-8 and VEGF. It is also related to a BBB structure alteration, with a decrease in tight-junction proteins such as zona occludens or claudin-5. Moreover, the BBB function reveals increased permeability and leukocyte adhesion in cells expressing SSAO/VAP-1, as well as an enhancement of the vascular Aβ deposition induced by mechanisms both dependent and independent of the enzymatic activity of SSAO/VAP-1. These results reveal an interesting role of vascular SSAO/VAP-1 in BBB dysfunction related to AD progression, opening a new window in the search of alternative therapeutic targets for fighting AD.
Collapse
Affiliation(s)
- Montse Solé
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain.
| | - María Esteban-Lopez
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Biel Taltavull
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Cristina Fábregas
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Rut Fadó
- Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Núria Casals
- Basic Sciences Department, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Santiago de Compostela, Spain
| | - Jose Rodríguez-Álvarez
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Alfredo J Miñano-Molina
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Mercedes Unzeta
- Biochemistry and Molecular Biology Department, Institute of Neurosciences (INc), Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain.
| |
Collapse
|
134
|
Farfara D, Feierman E, Richards A, Revenko AS, MacLeod RA, Norris EH, Strickland S. Knockdown of circulating C1 inhibitor induces neurovascular impairment, glial cell activation, neuroinflammation, and behavioral deficits. Glia 2019; 67:1359-1373. [PMID: 30882931 DOI: 10.1002/glia.23611] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 02/11/2019] [Accepted: 02/19/2019] [Indexed: 12/20/2022]
Abstract
The cross-talk between blood proteins, immune cells, and brain function involves complex mechanisms. Plasma protein C1 inhibitor (C1INH) is an inhibitor of vascular inflammation that is induced by activation of the kallikrein-kinin system (KKS) and the complement system. Knockout of C1INH was previously correlated with peripheral vascular permeability via the bradykinin pathway, yet there was no evidence of its correlation with blood-brain barrier (BBB) integrity and brain function. In order to understand the effect of plasma C1INH on brain pathology via the vascular system, we knocked down circulating C1INH in wild-type (WT) mice using an antisense oligonucleotide (ASO), without affecting C1INH expression in peripheral immune cells or the brain, and examined brain pathology. Long-term elimination of endogenous C1INH in the plasma induced the activation of the KKS and peritoneal macrophages but did not activate the complement system. Bradykinin pathway proteins were elevated in the periphery and the brain, resulting in hypotension. BBB permeability, extravasation of plasma proteins into the brain parenchyma, activation of glial cells, and elevation of pro-inflammatory response mediators were detected. Furthermore, infiltrating innate immune cells were observed entering the brain through the lateral ventricle walls and the neurovascular unit. Mice showed normal locomotion function, yet cognition was impaired and depressive-like behavior was evident. In conclusion, our results highlight the important role of regulated plasma C1INH as it acts as a gatekeeper to the brain via the neurovascular system. Thus, manipulation of C1INH in neurovascular disorders might be therapeutically beneficial.
Collapse
Affiliation(s)
- Dorit Farfara
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| | - Emily Feierman
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| | - Allison Richards
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| | - Alexey S Revenko
- Department of Antisense Drug Discovery, IONIS Pharmaceuticals Inc., Carlsbad, California
| | - Robert A MacLeod
- Department of Antisense Drug Discovery, IONIS Pharmaceuticals Inc., Carlsbad, California
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| |
Collapse
|
135
|
Tangestani Fard M, Stough C. A Review and Hypothesized Model of the Mechanisms That Underpin the Relationship Between Inflammation and Cognition in the Elderly. Front Aging Neurosci 2019; 11:56. [PMID: 30930767 PMCID: PMC6425084 DOI: 10.3389/fnagi.2019.00056] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/26/2019] [Indexed: 12/13/2022] Open
Abstract
Age is associated with increased risk for several disorders including dementias, cardiovascular disease, atherosclerosis, obesity, and diabetes. Age is also associated with cognitive decline particularly in cognitive domains associated with memory and processing speed. With increasing life expectancies in many countries, the number of people experiencing age-associated cognitive impairment is increasing and therefore from both economic and social terms the amelioration or slowing of cognitive aging is an important target for future research. However, the biological causes of age associated cognitive decline are not yet, well understood. In the current review, we outline the role of inflammation in cognitive aging and describe the role of several inflammatory processes, including inflamm-aging, vascular inflammation, and neuroinflammation which have both direct effect on brain function and indirect effects on brain function via changes in cardiovascular function.
Collapse
Affiliation(s)
| | - Con Stough
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|
136
|
Bettcher BM, Neuhaus J, Wynn MJ, Elahi FM, Casaletto KB, Saloner R, Fitch R, Karydas A, Kramer JH. Increases in a Pro-inflammatory Chemokine, MCP-1, Are Related to Decreases in Memory Over Time. Front Aging Neurosci 2019; 11:25. [PMID: 30814948 PMCID: PMC6381047 DOI: 10.3389/fnagi.2019.00025] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/29/2019] [Indexed: 12/28/2022] Open
Abstract
Objective: To determine the longitudinal relationship between monocyte chemotactic protein 1 (MCP-1)/CCL2 and memory function in older adults. Methods: We examined longitudinal plasma MCP-1/CCL2 levels and a longitudinal verbal memory measure (CVLT-II 20' recall) in a sample of 399 asymptomatic older adults (mean age = 72.1). Total visits ranged from 1 to 8, with an average time of 2.1 years between each visit, yielding 932 total observations. In order to isolate change over time, we decomposed MCP-1/CCL2 into subject-specific means and longitudinal deviations from the mean. The decomposed MCP-1/CCL2 variables were entered as predictors in linear mixed effects models, with age at baseline, sex, and education entered as covariates and recall as the longitudinal outcome. In follow-up analyses, we controlled for global cognition and APOE genotype, as well as baseline vascular risk factors. We also examined the specificity of findings by examining the longitudinal association between the MCP-1/CCL2 variables and non-memory cognitive tests. Results: Within-subject increases in MCP-1/CCL2 levels were associated with decreases in delayed recall (t = -2.65; p = 0.01) over time. Results were independent of global cognitive function and APOE status (t = -2.30, p = 0.02), and effects remained when controlling for baseline vascular risk factors (t = -1.92, p = 0.05). No associations were noted between within-subject increases in MCP-1/CCL2 levels and other cognitive domains. Conclusions: In an asymptomatic aging adult cohort, longitudinal increases in MCP-1/CCL2 levels were associated with longitudinal decline in memory. Results suggest that "healthy aging" is typified by early remodeling of the immune system, and that the chemokine, MCP-1/CCL2, may be associated with negative memory outcomes.
Collapse
Affiliation(s)
- Brianne M Bettcher
- Rocky Mountain Alzheimer's Disease Center, Departments of Neurosurgery and Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - John Neuhaus
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Matthew J Wynn
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, United States
| | - Fanny M Elahi
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Kaitlin B Casaletto
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Rowan Saloner
- Department of Psychiatry, San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, United States
- HIV Neurobehavioral Research Program, Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Ryan Fitch
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Anna Karydas
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Joel H Kramer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
137
|
Festoff BW, Citron BA. Thrombin and the Coag-Inflammatory Nexus in Neurotrauma, ALS, and Other Neurodegenerative Disorders. Front Neurol 2019; 10:59. [PMID: 30804878 PMCID: PMC6371052 DOI: 10.3389/fneur.2019.00059] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
This review details our current understanding of thrombin signaling in neurodegeneration, with a focus on amyotrophic lateral sclerosis (ALS, Lou Gehrig's disease) as well as future directions to be pursued. The key factors are multifunctional and involved in regulatory pathways, namely innate immune and the coagulation cascade activation, that are essential for normal nervous system function and health. These two major host defense systems have a long history in evolution and include elements and regulators of the coagulation pathway that have significant impacts on both the peripheral and central nervous system in health and disease. The clotting cascade responds to a variety of insults to the CNS including injury and infection. The blood brain barrier is affected by these responses and its compromise also contributes to these detrimental effects. Important molecules in signaling that contribute to or protect against neurodegeneration include thrombin, thrombomodulin (TM), protease activated receptor 1 (PAR1), damage associated molecular patterns (DAMPs), such as high mobility group box protein 1 (HMGB1) and those released from mitochondria (mtDAMPs). Each of these molecules are entangled in choices dependent upon specific signaling pathways in play. For example, the particular cleavage of PAR1 by thrombin vs. activated protein C (APC) will have downstream effects through coupled factors to result in toxicity or neuroprotection. Furthermore, numerous interactions influence these choices such as the interplay between HMGB1, thrombin, and TM. Our hope is that improved understanding of the ways that components of the coagulation cascade affect innate immune inflammatory responses and influence the course of neurodegeneration, especially after injury, will lead to effective therapeutic approaches for ALS, traumatic brain injury, and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Barry W Festoff
- pHLOGISTIX LLC, Fairway, KS, United States.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Bruce A Citron
- Laboratory of Molecular Biology Research & Development, VA New Jersey Health Care System, East Orange, NJ, United States.,Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
138
|
de la Monte SM. The Full Spectrum of Alzheimer's Disease Is Rooted in Metabolic Derangements That Drive Type 3 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:45-83. [PMID: 31062325 PMCID: PMC9996398 DOI: 10.1007/978-981-13-3540-2_4] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The standard practice in neuropathology is to diagnose Alzheimer's disease (AD) based on the distribution and abundance of neurofibrillary tangles and Aβ deposits. However, other significant abnormalities including neuroinflammation, gliosis, white matter degeneration, non-Aβ microvascular disease, and insulin-related metabolic dysfunction require further study to understand how they could be targeted to more effectively remediate AD. This review addresses non-Aβ and non-pTau AD-associated pathologies, highlighting their major features, roles in neurodegeneration, and etiopathic links to deficits in brain insulin and insulin-like growth factor signaling and cognitive impairment. The discussion delineates why AD with its most characteristic clinical and pathological phenotypic profiles should be regarded as a brain form of diabetes, i.e., type 3 diabetes, and entertains the hypothesis that type 3 diabetes is just one of the categories of insulin resistance diseases that can occur independently or overlap with one or more of the others, including type 2 diabetes, metabolic syndrome, and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Departments of Neurology, Neuropathology, and Neurosurgery, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Pathology and Laboratory Medicine, Providence VA Medical Center, Providence, RI, USA.
| |
Collapse
|
139
|
Ranjan R, Rout M, Mishra M, Kore SA. Tooth loss and dementia: An oro-neural connection. A cross-sectional study. J Indian Soc Periodontol 2019; 23:158-162. [PMID: 30983788 PMCID: PMC6434721 DOI: 10.4103/jisp.jisp_430_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background: Recent cross-sectional studies have suggested that chronic inflammation resulting due to gingival and periodontal diseases may have potential reversible risk factors for dementia. Recently, cognition, memory deficit, and along with the behavioral defect have been considered as characteristics of dementia. Age is one of the main factors which is also known to be associated with dementia. The aim of the present study was to establish the correlation between tooth loss and diagnosis of cognitive impairment/dementia in elderly patients. Materials and Methods: A cross-sectional study, comprising of 300 patients (Age: 50–80 years), was conducted. For the assessment of cognitive impairment/dementia, Mini-Mental Status Examination (MMSE) was conducted. All the results were analyzed using the Chi-square and Kruskal–Wallis ANOVA test. Results: Number of teeth was found to be directly associated with MMSE. Less number of teeth was associated with lower MMSE score, irrespective of gender. The level of statistical significance was found to be P < 0.001. Factors, such as socioeconomic status, school education, and marital status, were found to have a statistically significant impact on the dementia scale. Conclusion: The current study found a significant association between tooth loss and cognitive impairment. Along with age factors such as socioeconomic status, education, and marital status were also found to have a direct or indirect impact on dementia. More prospective studies and clinical trials are required to be conducted to confirm the observations of this study.
Collapse
Affiliation(s)
- Rajeev Ranjan
- Community Health Centre, Government of Jharkhand, Bero-Ranchi, Jharkhand, India
| | - Manisha Rout
- Kalinga Institute of Dental Sciences, KIIT University, Bhubaneswar, Odisha, India
| | - Monalisa Mishra
- Department of Life Science, NIT Rourkela, Rourkela, Odisha, India
| | - Shobha Abhijeet Kore
- Smile Care Dental Clinic and Implant Center, Shaniwar Peth, Karad, Maharashtra, India
| |
Collapse
|
140
|
|
141
|
Occupational exposure to extremely low frequency magnetic fields and risk of Alzheimer disease: A systematic review and meta-analysis. Neurotoxicology 2018; 69:242-252. [DOI: 10.1016/j.neuro.2017.12.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 12/16/2022]
|
142
|
Dementia and Atrial Fibrillation: Pathophysiological Mechanisms and Therapeutic Implications. Am J Med 2018; 131:1408-1417. [PMID: 30076825 DOI: 10.1016/j.amjmed.2018.06.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 11/23/2022]
Abstract
Atrial fibrillation increases the risk of stroke by a factor of four- to fivefold, and dementia is a common consequence of stroke. However, atrial fibrillation has been associated with cognitive impairment and dementia, even in patients without prior overt stroke. Nonischemic mechanisms include cerebral hypoperfusion, vascular inflammation, brain atrophy, genetic factors, and shared risk factors such as age or hypertension. Critical appraisal of studies evaluating the association between atrial fibrillation and dementia in stroke-free patients reveals that several suffer from methodological issues, such as not including silent stroke or anticoagulation therapy in multivariate analyses. Some studies show a close relationship between atrial fibrillation and dementia due to silent stroke, in the absence of overt stroke. Evidence is accumulating that anticoagulation may be effective to decrease the risk of dementia in atrial fibrillation patients. Overall, the pathogenesis linking atrial fibrillation to dementia is likely multifactorial. Cerebral infarctions, including silent stroke, play a central role. These findings underscore the importance of stroke prevention measures in atrial fibrillation patients.
Collapse
|
143
|
de J.R. De-Paula V, Forlenza AS, Forlenza OV. Relevance of gutmicrobiota in cognition, behaviour and Alzheimer’s disease. Pharmacol Res 2018; 136:29-34. [DOI: 10.1016/j.phrs.2018.07.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/30/2018] [Accepted: 07/10/2018] [Indexed: 12/22/2022]
|
144
|
Khan MM, Xiao J, Patel D, LeDoux MS. DNA damage and neurodegenerative phenotypes in aged Ciz1 null mice. Neurobiol Aging 2018; 62:180-190. [PMID: 29154038 DOI: 10.1016/j.neurobiolaging.2017.10.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/17/2017] [Accepted: 10/16/2017] [Indexed: 12/28/2022]
Abstract
Cell-cycle dysfunction and faulty DNA repair are closely intertwined pathobiological processes that may contribute to several neurodegenerative disorders. CDKN1A interacting zinc finger protein 1 (CIZ1) plays a critical role in DNA replication and cell-cycle progression at the G1/S checkpoint. Germline or somatic variants in CIZ1 have been linked to several neural and extra-neural diseases. Recently, we showed that germline knockout of Ciz1 is associated with motor and hematological abnormalities in young adult mice. However, the effects of CIZ1 deficiency in much older mice may be more relevant to understanding age-related declines in cognitive and motor functioning and age-related neurologic disorders such as isolated dystonia and Alzheimer disease. Mouse embryonic fibroblasts from Ciz1-/- mice showed abnormal sensitivity to the effects of γ-irradiation with persistent DNA breaks, aberrant cell-cycle progression, and apoptosis. Aged (18-month-old) Ciz1-/- mice exhibited marked deficits in motor and cognitive functioning, and, in brain tissues, overt DNA damage, NF-κB upregulation, oxidative stress, vascular dysfunction, inflammation, and cell death. These findings indicate that the deleterious effects of CIZ1 deficiency become more pronounced with aging and suggest that defects of cell-cycle control and associated DNA repair pathways in postmitotic neurons could contribute to global neurologic decline in elderly human populations. Accordingly, the G1/S cell-cycle checkpoint and associated DNA repair pathways may be targets for the prevention and treatment of age-related neurodegenerative processes.
Collapse
Affiliation(s)
- Mohammad Moshahid Khan
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jianfeng Xiao
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Damini Patel
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mark S LeDoux
- Departments of Neurology, and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
145
|
Kinney JW, Bemiller SM, Murtishaw AS, Leisgang AM, Salazar AM, Lamb BT. Inflammation as a central mechanism in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2018; 4:575-590. [PMID: 30406177 PMCID: PMC6214864 DOI: 10.1016/j.trci.2018.06.014] [Citation(s) in RCA: 1366] [Impact Index Per Article: 195.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by cognitive decline and the presence of two core pathologies, amyloid β plaques and neurofibrillary tangles. Over the last decade, the presence of a sustained immune response in the brain has emerged as a third core pathology in AD. The sustained activation of the brain's resident macrophages (microglia) and other immune cells has been demonstrated to exacerbate both amyloid and tau pathology and may serve as a link in the pathogenesis of the disorder. In the following review, we provide an overview of inflammation in AD and a detailed coverage of a number of microglia-related signaling mechanisms that have been implicated in AD. Additional information on microglia signaling and a number of cytokines in AD are also reviewed. We also review the potential connection of risk factors for AD and how they may be related to inflammatory mechanisms.
Collapse
Affiliation(s)
- Jefferson W. Kinney
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Shane M. Bemiller
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew S. Murtishaw
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Amanda M. Leisgang
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Arnold M. Salazar
- Department of Psychology, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Bruce T. Lamb
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
146
|
Freed JK, Durand MJ. There is no way to sugar coat it, you are getting older. Am J Physiol Heart Circ Physiol 2018; 315:H632-H633. [PMID: 29906230 DOI: 10.1152/ajpheart.00313.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Julie K Freed
- Department of Anesthesiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Matthew J Durand
- Department of Physical Medicine and Rehabilitation, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
147
|
Valacchi G, Virgili F, Cervellati C, Pecorelli A. OxInflammation: From Subclinical Condition to Pathological Biomarker. Front Physiol 2018; 9:858. [PMID: 30038581 PMCID: PMC6046448 DOI: 10.3389/fphys.2018.00858] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/15/2018] [Indexed: 12/18/2022] Open
Abstract
Inflammation is a complex systemic response evolved to cope with cellular injury, either due to infectious agents or, in general, with sporadic events challenging tissue integrity and function. Researchers involved in different fields have the tendency to look at the inflammatory response with different angles, according to their specific interest. Established its complexity, one of the most evident features of the inflammatory response is the generation of a pro-oxidative environment due to the production of high fluxes of pro-oxidant species. This production begins locally, close to the sites of tissue damage or infection, but eventually becomes a chronic challenge for the organism, if the inflammatory response is not properly controlled. In this review, we focus on this specific aspect of chronic, low-level sub-clinical inflammatory response. We propose the term "OxInflammation" as a novel operative term describing a permanent pro-oxidative feature that interact, in a positive feed-back manner, to a not yet clinically detectable inflammatory process, leading in a long run (chronically) to a systemic/local damage, as a consequence of the cross talk between inflammatory, and oxidative stress mediators. Therefore, it could be useful to analyze inflammatory markers in pathologies where there is an alteration of the redox homeostasis, although an inflammatory status is not clinically evident.
Collapse
Affiliation(s)
- Giuseppe Valacchi
- Plants for Human Health Institute, Department of Animal Sciences, North Carolina State University, Kannapolis, NC, United States
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Fabio Virgili
- Council for Agricultural Research and Economics - Food and Nutrition Research Centre (C.R.E.A.-AN), Rome, Italy
| | - Carlo Cervellati
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandra Pecorelli
- Plants for Human Health Institute, Department of Animal Sciences, North Carolina State University, Kannapolis, NC, United States
| |
Collapse
|
148
|
Bevelacqua JJ, Mortazavi SMJ. Alzheimer 's Disease: Possible Mechanisms Behind Neurohormesis Induced by Exposure to Low Doses of Ionizing Radiation. J Biomed Phys Eng 2018; 8:153-156. [PMID: 29951441 PMCID: PMC6015644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/02/2018] [Indexed: 06/08/2023]
Abstract
In 2016, scientists reported that human exposure to low doses of ionizing radiation (CT scans of the brain) might relieve symptoms of both Alzheimer's disease (AD) and Parkinson disease (PD). The findings were unbelievable for those who were not familiar with neurohormesis. X-ray stimulation of the patient's adaptive protection systems against neurodegenerative diseases was the mechanism proposed by those authors. Now, some more recent studies performed in the field of neurobiological research confirm that low levels of stress can produce protective responses against the pathogenic processes. This paper outlines possible protective consequences of LDR in preventing the pathogenesis of AD through mechanisms such as restoring the myelin sheath and preventing neurodegeneration caused by oxidative stress. Focal demyelination is frequently reported in the proximity of beta-amyloid plaques within neocortex. Extracellular accumulation of amyloid is among well-characterized pathological changes in AD. It should be noted that LDR has been shown to contribute to the regeneration and functional recovery after transverse peripheral nerve injury (through inducing increased production of VEGF and GAP-43), which advances both the axonal regeneration and myelination. Another mechanism which is possibly involved is preventing neurodegeneration caused by oxidative stress. While high doses can induce reactive oxygen species (ROS) formation, oxidative stress and neuro-inflammation, substantial evidence now indicates that LDR can mitigate tissue damage through antioxidant defenses. Although adult neurogenesis has been reported to be beneficial for the regeneration of nervous system, some studies demonstrate that neurogenesis increases in AD brains. In spite of these reports, cellular therapy is introduced as a promising strategy for AD, and hence, LDR can affect the proliferation and differentiation of neural stem cells. Although such mechanisms are not fully known yet, it is hoped that this paper would foster further investigation into the mechanisms of this phenomenon, which accordingly improves human health.
Collapse
Affiliation(s)
- J J Bevelacqua
- Bevelacqua Resources, 343 Adair Drive, Richland, WA 99352, USA
| | - S M J Mortazavi
- Biophotonics Lab, Department of Electrical Engineering, University of Wisconsin Milwaukee, 3200 N Cramer St., Milwaukee, WI 53211, USA
- Ionizing and Non-ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
149
|
Sen A, Hongpaisan J. Hippocampal microvasculature changes in association with oxidative stress in Alzheimer's disease. Free Radic Biol Med 2018; 120:192-203. [PMID: 29572097 DOI: 10.1016/j.freeradbiomed.2018.03.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 03/14/2018] [Accepted: 03/17/2018] [Indexed: 12/13/2022]
Abstract
Vascular endothelial dysfunction is a primary phenotype of aging, and microvascular (MV) lesion is mainly associated with Alzheimer's disease (AD). Here we have studied the correlation of MV wall thickness and CA1 pyramidal neuronal pathology in autopsy-confirmed AD brains. Both hyaline (h-MV) and increased cell number (c-MV) associated MV wall thickening was found in age-matched control (AC) hippocampus without significant change in Aβ level (Braak stages 0-III). AC neurons neighboring the h-MV showed lower levels of oxidative DNA/RNA damage and Aβ precursor protein (APP), while the neurons around c-MV showed higher oxidative DNA/RNA damage with increased APP expression. Neurons in AC hippocampus without MV wall thickening (thin wall) showed increased DNA/RNA damage and APP levels compared to AC cases with h-MV and c-MV walls. In the AD hippocampus neurons neighboring h-MV walls showed increased levels of Aβ and decreased number of dendritic spines (at Braak stages IV-VI). C-MV neighboring neurons in the AD cases showed higher levels of DNA/RNA damage with increased APP at stages II - III, followed by lower levels of oxidative DNA/RNA damage, decreased APP and increased Aβ levels with loss of dendritic spines at stages IV-VI. Prolonged treatment of primary human fetal hippocampal neurons with tert-butyl hydroperoxide (TBHP) induced oxidative DNA damage with a sustained increase in APP. Aβ increased rapidly and then decreased overtime. Short-term TBHP treated neurons showed lower levels of superoxide (O2• -) without significant DNA damage. Short-term TBHP treatment induced a gradual decrease in APP but an increase in Aβ levels over time. In conclusion this study indicates that AD hippocampus at Braak stages II-III are characterized by strong oxidative DNA/RNA damage with increased APP in neurons associated with c-MV, while stages IV-VI are characterized by a slow increase in Aβ in neurons neighboring both h-MV and c-MV.
Collapse
Affiliation(s)
- Abhik Sen
- Center for Neurodegenerative Diseases, Blanchette Rockefeller Neurosciences Institute, West Virginia University, Morgantown, WV, 26505, USA
| | - Jarin Hongpaisan
- Center for Neurodegenerative Diseases, Blanchette Rockefeller Neurosciences Institute, West Virginia University, Morgantown, WV, 26505, USA.
| |
Collapse
|
150
|
Smyth LCD, Rustenhoven J, Park TIH, Schweder P, Jansson D, Heppner PA, O'Carroll SJ, Mee EW, Faull RLM, Curtis M, Dragunow M. Unique and shared inflammatory profiles of human brain endothelia and pericytes. J Neuroinflammation 2018; 15:138. [PMID: 29751771 PMCID: PMC5948925 DOI: 10.1186/s12974-018-1167-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/18/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Pericytes and endothelial cells are critical cellular components of the blood-brain barrier (BBB) and play an important role in neuroinflammation. To date, the majority of inflammation-related studies in endothelia and pericytes have been carried out using immortalised cell lines or non-human-derived cells. Whether these are representative of primary human cells is unclear and systematic comparisons of the inflammatory responses of primary human brain-derived pericytes and endothelia has yet to be performed. METHODS To study the effects of neuroinflammation at the BBB, primary brain endothelial cells and pericytes were isolated from human biopsy tissue. Culture purity was examined using qPCR and immunocytochemistry. Electrical cell-substrate impedance sensing (ECIS) was used to determine the barrier properties of endothelial and pericyte cultures. Using immunocytochemistry, cytometric bead array, and ECIS, we compared the responses of endothelia and pericytes to a panel of inflammatory stimuli (IL-1β, TNFα, LPS, IFN-γ, TGF-β1, IL-6, and IL-4). Secretome analysis was performed to identify unique secretions of endothelia and pericytes in response to IL-1β. RESULTS Endothelial cells were pure, moderately proliferative, retained the expression of BBB-related junctional proteins and transporters, and generated robust TEER. Both endothelia and pericytes have the same pattern of transcription factor activation in response to inflammatory stimuli but respond differently at the secretion level. Secretome analysis confirmed that endothelia and pericytes have overlapping but distinct secretome profiles in response to IL-1β. We identified several cell-type specific responses, including G-CSF and GM-CSF (endothelial-specific), and IGFBP2 and IGFBP3 (pericyte-specific). Finally, we demonstrated that direct addition of IL-1β, TNFα, LPS, and IL-4 contributed to the loss of endothelial barrier integrity in vitro. CONCLUSIONS Here, we identify important cell-type differences in the inflammatory response of brain pericytes and endothelia and provide, for the first time, a comprehensive profile of the secretions of primary human brain endothelia and pericytes which has implications for understanding how inflammation affects the cerebrovasculature.
Collapse
Affiliation(s)
- Leon C D Smyth
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Thomas I-H Park
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Patrick Schweder
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Deidre Jansson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Peter A Heppner
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Simon J O'Carroll
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Edward W Mee
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Maurice Curtis
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand. .,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|