1
|
Yadav S, Deepika, Moar K, Kumar A, Khola N, Pant A, Kakde GS, Maurya PK. Reconsidering red blood cells as the diagnostic potential for neurodegenerative disorders. Biol Cell 2024; 116:e2400019. [PMID: 38822416 DOI: 10.1111/boc.202400019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/12/2024] [Accepted: 04/29/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Red blood cells (RBCs) are usually considered simple cells and transporters of gases to tissues. HYPOTHESIS However, recent research has suggested that RBCs may have diagnostic potential in major neurodegenerative disorders (NDDs). RESULTS This review summarizes the current knowledge on changes in RBC in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and other NDDs. It discusses the deposition of neuronal proteins like amyloid-β, tau, and α-synuclein, polyamines, changes in the proteins of RBCs like band-3, membrane transporter proteins, heat shock proteins, oxidative stress biomarkers, and altered metabolic pathways in RBCs during neurodegeneration. It also highlights the comparison of RBC diagnostic markers to other in-market diagnoses and discusses the challenges in utilizing RBCs as diagnostic tools, such as the need for standardized protocols and further validation studies. SIGNIFICANCE STATEMENT The evidence suggests that RBCs have diagnostic potential in neurodegenerative disorders, and this study can pave the foundation for further research which may lead to the development of novel diagnostic approaches and treatments.
Collapse
Affiliation(s)
- Somu Yadav
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Deepika
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Kareena Moar
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Akshay Kumar
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Nikhila Khola
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Anuja Pant
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Ganseh S Kakde
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh, India
| |
Collapse
|
2
|
Lymperopoulos D, Dedemadi AG, Voulgari ML, Georgiou E, Dafnis I, Mountaki C, Panagopoulou EA, Karvelas M, Chiou A, Karathanos VT, Chroni A. Corinthian Currants Promote the Expression of Paraoxonase-1 and Enhance the Antioxidant Status in Serum and Brain of 5xFAD Mouse Model of Alzheimer's Disease. Biomolecules 2024; 14:426. [PMID: 38672443 PMCID: PMC11047902 DOI: 10.3390/biom14040426] [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: 03/07/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Paraoxonase-1 (PON1), a serum antioxidant enzyme, has been implicated in Alzheimer's disease (AD) pathogenesis that involves early oxidative damage. Corinthian currants and their components have been shown to display antioxidant and other neuroprotective effects in AD. We evaluated the effect of a Corinthian currant paste-supplemented diet (CurD), provided to 1-month-old 5xFAD mice for 1, 3, and 6 months, on PON1 activity and levels of oxidation markers in serum and the brain of mice as compared to a control diet (ConD) or glucose/fructose-matched diet (GFD). Administration of CurD for 1 month increased PON1 activity and decreased oxidized lipid levels in serum compared to ConD and GFD. Longer-term administration of CurD did not, however, affect serum PON1 activity and oxidized lipid levels. Furthermore, CurD administered for 1 and 3 months, but not for 6 months, increased PON1 activity and decreased free radical levels in the cortex of mice compared to ConD and GFD. To probe the mechanism for the increased PON1 activity in mice, we studied the effect of Corinthian currant polar phenolic extract on PON1 activity secreted by Huh-7 hepatocytes or HEK293 cells transfected with a PON1-expressing plasmid. Incubation of cells with the extract led to a dose-dependent increase of secreted PON1 activity, which was attributed to increased cellular PON1 expression. Collectively, our findings suggest that phenolics in Corinthian currants can increase the hepatic expression and activity of antioxidant enzyme PON1 and that a Corinthian currant-supplemented diet during the early stages of AD in mice reduces brain oxidative stress.
Collapse
Affiliation(s)
- Dimitris Lymperopoulos
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
- Department of Biology, National and Kapodistrian University of Athens, Zografou, 15784 Athens, Greece
| | - Anastasia-Georgia Dedemadi
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
- Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15784 Athens, Greece
| | - Maria-Lydia Voulgari
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
- Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15784 Athens, Greece
| | - Eirini Georgiou
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
- Department of Chemistry, National and Kapodistrian University of Athens, Zografou, 15784 Athens, Greece
| | - Ioannis Dafnis
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
| | - Christina Mountaki
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
| | - Eirini A. Panagopoulou
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Dietetics and Nutrition, Harokopio University, 17676 Kallithea, Greece (A.C.); (V.T.K.)
| | - Michalis Karvelas
- Research and Development Department, Agricultural Cooperatives’ Union of Aeghion, 25100 Aeghion, Greece;
| | - Antonia Chiou
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Dietetics and Nutrition, Harokopio University, 17676 Kallithea, Greece (A.C.); (V.T.K.)
| | - Vaios T. Karathanos
- Laboratory of Chemistry-Biochemistry-Physical Chemistry of Foods, Department of Dietetics and Nutrition, Harokopio University, 17676 Kallithea, Greece (A.C.); (V.T.K.)
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, 15341 Athens, Greece
| |
Collapse
|
3
|
Xia X, Chen K, Chen Y. Change in function and homeostasis of HPA axis: The role of vitamin family. Chem Biol Interact 2024; 391:110899. [PMID: 38325521 DOI: 10.1016/j.cbi.2024.110899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/24/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
With the improvement of living quality, people pay more and more attention to vitamin supplements. The vitamins in the daily diet can meet the needs of the body. Whether additional vitamin supplementation is necessary still needs to be further explored. Many studies have reported that vitamin deficiency and excessive vitamin supplementation could lead to abnormal development in the body or increase the risk of diseases. Here, we summarize the abnormal levels of vitamins can cause the homeostasis imbalance of hypothalamus-pituitary-adrenal (HPA) axis by affecting its development and function. It can lead to abnormal synthesis and secretion of glucocorticoid in the body, which mediates the occurrence and development of metabolic diseases and psychoneurotic diseases. In addition, vitamin has a strong antioxidant effect, which can eliminate oxygen free radicals. Thereby, vitamins can alter HPA axis function and homeostasis maintenance by combating oxidative stress. This review provides a theoretical basis for clarifying the role of abnormal levels of vitamin in the occurrence and development of multiple diseases and its intervention strategy, and also provides reference value and guiding significance for rational use of vitamins.
Collapse
Affiliation(s)
- Xuan Xia
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kaiqi Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yawen Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
4
|
Zeinalabedini M, Mousavi Z, Amjadi A, Shapouri M, Aminnezhad Kavkani B, Masoumvand M, Mobarakeh KA, Gholamalizadeh M, Valisoltani N, Mohammadi S, Khoshdooz S, Doaei S, Kooshki A. Does dietary intake of caffeine have an effect on transient global amnesia? Neuropsychopharmacol Rep 2024; 44:143-148. [PMID: 38131259 PMCID: PMC10932796 DOI: 10.1002/npr2.12408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
AIM Amnesia is a cognitive disorder that may lead to memory loss. Caffeine is a psychoactive substance which have an effect on memory and cognitive functions. This study aimed to assess the association of transient global amnesia (TGA) with dietary intake of caffeine. METHODS This cross-sectional study was conducted on the Sabzevar Persian cohort data of 258 patients with TGA and 520 healthy individuals in Sabzevar, Iran. The Nutritional data were gathered in face-to-face interviews using a valid Food Frequency Questionnaire. Different models of logistic regression were used to determine the association between TGA and dietary caffeine intake after adjusting the confounders including age, sex, education, job, marital status, physical activity, BMI, and calorie intake. RESULTS There was no significant difference in terms of dietary calorie intake of (2279.5 ± 757.9 vs. 2365.5 ± 799.5, p = 0.19), protein (70.79 ± 25.27 vs. 72.94 ± 24.83, p = 0.31), fat (59.97 ± 23.79 vs. 60.13 ± 26.38, p = 0.93), carbohydrate (376 ± 134 vs. 393.1 ± 137.8, p = 0.14), and caffeine (196.4 ± 127.9 vs. 186.3 ± 128.5, p = 0.36) between the groups. No significant association was found between TGA and dietary intake of caffeine (OR: 0.99, 95% CI: 0.99-1.01, p = 0.36). The results did not change after adjusting the confounders. CONCLUSIONS No significant association was found between TGA and dietary intake of caffeine. Further prospective studies are required to confirm this finding.
Collapse
Affiliation(s)
- Mobina Zeinalabedini
- Department of Community of Nutrition, School of Nutritional Sciences and DieteticTehran University of Medical SciencesTehranIran
| | - Zahra Mousavi
- School of Nursing and MidwiferyShahed UniversityTehranIran
| | - Arezoo Amjadi
- Department of Nutrition, School of Nutritional Sciences and Food TechnologyKermanshah University of Medical SciencesKermanshahIran
| | - Mahsa Shapouri
- Shahid Beheshti University of Medical SciencesTehranIran
| | | | - Mohammad Masoumvand
- Department of Nutrition, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Khadijeh Abbasi Mobarakeh
- Department of Community Nutrition, Nutrition and Food Security Research Center, School of Nutrition and Food ScienceIsfahan University of Medical SciencesIsfahanIran
| | | | - Neda Valisoltani
- Department of Clinical Nutrition, School of Nutritional Sciences and DieteticsTehran University of Medical SciencesTehranIran
| | - Saeideh Mohammadi
- Department of NutritionZanjan University of Medical SciencesZanjanIran
| | - Sara Khoshdooz
- Faculty of MedicineGuilan University of Medical SciencesRashtIran
| | - Saeid Doaei
- Department of Community Nutrition, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food TechnologyShahid Beheshti University of Medical SciencesTehranIran
| | - Akram Kooshki
- Non‐Communicable Diseases Research Center, Department of Nutrition & Biochemistry, Faculty of MedicineSabzevar University of Medical SciencesSabzevarIran
| |
Collapse
|
5
|
Nasb M, Tao W, Chen N. Alzheimer's Disease Puzzle: Delving into Pathogenesis Hypotheses. Aging Dis 2024; 15:43-73. [PMID: 37450931 PMCID: PMC10796101 DOI: 10.14336/ad.2023.0608] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disease characterized by both amnestic and non-amnestic clinical manifestations. It accounts for approximately 60-70% of all dementia cases worldwide. With the increasing number of AD patients, elucidating underlying mechanisms and developing corresponding interventional strategies are necessary. Hypotheses about AD such as amyloid cascade, Tau hyper-phosphorylation, neuroinflammation, oxidative stress, mitochondrial dysfunction, cholinergic, and vascular hypotheses are not mutually exclusive, and all of them play a certain role in the development of AD. The amyloid cascade hypothesis is currently the most widely studied; however, other hypotheses are also gaining support. This article summarizes the recent evidence regarding major pathological hypotheses of AD and their potential interplay, as well as the strengths and weaknesses of each hypothesis and their implications for the development of effective treatments. This could stimulate further studies and promote the development of more effective therapeutic strategies for AD.
Collapse
Affiliation(s)
| | | | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| |
Collapse
|
6
|
Li T, Bao X, Li L, Qin R, Li C, Wang X. Heart failure and cognitive impairment: A narrative review of neuroimaging mechanism from the perspective of brain MRI. Front Neurosci 2023; 17:1148400. [PMID: 37051150 PMCID: PMC10083289 DOI: 10.3389/fnins.2023.1148400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/01/2023] [Indexed: 03/28/2023] Open
Abstract
Both heart failure (HF) and cognitive impairment (CI) have a significant negative impact on the health of the elderly individuals. Magnetic resonance imaging (MRI) can non-invasively detect functional and structural variations in the heart and brain, making it easier to explore the connection between the heart and brain. According to neuroimaging studies, HF patients have a higher chance of developing CI because they have a variety of different types of brain injuries. To examine how HF and CI are influenced by one another, English-language literature was searched in the Web of Science, PubMed EMBASE (OVID), PsycInfo, and Scopus databases. The search terms included “high-frequency,” “brain function,” “brain injury,” “cognition,” “cognitive impairment,” and “magnetic resonance imaging.” Normal brain function is typically impaired by HF in the form of decreased cerebral perfusion pressure, inflammation, oxidative stress, and damage to the BBB, resulting in CI and subsequent HF. Early pathophysiological alterations in patients’ brains have been widely detected using a range of novel MRI techniques, opening up new avenues for investigating the connection between HF and CI. This review aims to describe the pathogenesis of HF with CI and the early diagnostic role of MRI in the heart-brain domain.
Collapse
Affiliation(s)
- Tong Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangyuan Bao
- School of Radiology, Shandong First Medical University, Taian, China
| | - Lin Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rui Qin
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Cuicui Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Cuicui Li,
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Ximing Wang,
| |
Collapse
|
7
|
Iova OM, Marin GE, Lazar I, Stanescu I, Dogaru G, Nicula CA, Bulboacă AE. Nitric Oxide/Nitric Oxide Synthase System in the Pathogenesis of Neurodegenerative Disorders-An Overview. Antioxidants (Basel) 2023; 12:antiox12030753. [PMID: 36979000 PMCID: PMC10045816 DOI: 10.3390/antiox12030753] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/24/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Nitric oxide, a ubiquitous molecule found throughout the natural world, is a key molecule implicated in many central and benefic molecular pathways and has a well-established role in the function of the central nervous system, as numerous studies have previously shown. Dysregulation of its metabolism, mainly the upregulation of nitric oxide production, has been proposed as a trigger and/or aggravator for many neurological affections. Increasing evidence supports the implication of this molecule in prevalent neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, or amyotrophic lateral sclerosis. The mechanisms proposed for its neurotoxicity mainly center around the increased quantities of nitric oxide that are produced in the brain, their cause, and, most importantly, the pathological metabolic cascades created. These cascades lead to the formation of neuronal toxic substances that impair the neurons' function and structure on multiple levels. The purpose of this review is to present the main causes of increased pathological production, as well as the most important pathophysiological mechanisms triggered by nitric oxide, mechanisms that could help explain a part of the complex picture of neurodegenerative diseases and help develop targeted therapies.
Collapse
Affiliation(s)
- Olga-Maria Iova
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Gheorghe-Eduard Marin
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Izabella Lazar
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Stanescu
- Department of Neurology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Gabriela Dogaru
- Department of Physical Medicine and Rehabilitation, Iuliu Haţieganu University of Medicine and Pharmacy Cluj-Napoca, Viilor Street, No. 46-50, 400347 Cluj-Napoca, Romania
| | - Cristina Ariadna Nicula
- Department of Ophthalmology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Adriana Elena Bulboacă
- Department of Pathophysiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
8
|
Robin J, Demené C, Heiles B, Blanvillain V, Puke L, Perren F, Tanter M. In vivoadaptive focusing for clinical contrast-enhanced transcranial ultrasound imaging in human. Phys Med Biol 2023; 68. [PMID: 36595330 DOI: 10.1088/1361-6560/acabfb] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Objective. Imaging the human brain vasculature with high spatial and temporal resolution remains challenging in the clinic today. Transcranial ultrasound is still scarcely used for cerebrovascular imaging, due to low sensitivity and strong phase aberrations induced by the skull bone that only enable the proximal part major brain vessel imaging, even with ultrasound contrast agent injection (microbubbles).Approach. Here, we propose an adaptive aberration correction technique for skull bone aberrations based on the backscattered signals coming from intravenously injected microbubbles. Our aberration correction technique was implemented to image brain vasculature in human adults through temporal and occipital bone windows. For each subject, an effective speed of sound, as well as a phase aberration profile, were determined in several isoplanatic patches spread across the image. This information was then used in the beamforming process.Main results. This aberration correction method reduced the number of artefacts, such as ghost vessels, in the images. It improved image quality both for ultrafast Doppler imaging and ultrasound localization microscopy (ULM), especially in patients with thick bone windows. For ultrafast Doppler images, the contrast was increased by 4 dB on average, and for ULM, the number of detected microbubble tracks was increased by 38%.Significance. This technique is thus promising for better diagnosis and follow-up of brain pathologies such as aneurysms, arterial stenoses, arterial occlusions, microvascular disease and stroke and could make transcranial ultrasound imaging possible even in particularly difficult-to-image human adults.
Collapse
Affiliation(s)
- Justine Robin
- Physics for Medicine Paris, INSERM, ESPCI Paris, PSL Research University, CNRS, France.,LUNIC Laboratory, Neurocenter University of Geneva, Switzerland
| | - Charlie Demené
- Physics for Medicine Paris, INSERM, ESPCI Paris, PSL Research University, CNRS, France.,LUNIC Laboratory, Neurocenter University of Geneva, Switzerland
| | - Baptiste Heiles
- Physics for Medicine Paris, INSERM, ESPCI Paris, PSL Research University, CNRS, France
| | - Victor Blanvillain
- Physics for Medicine Paris, INSERM, ESPCI Paris, PSL Research University, CNRS, France
| | - Liene Puke
- LUNIC Laboratory, Neurocenter University of Geneva, Switzerland
| | - Fabienne Perren
- LUNIC Laboratory, Neurocenter University of Geneva, Switzerland.,Dept. of Clinical Neurosciences, Universities of Geneva and Fribourg, Switzerland
| | - Mickael Tanter
- Physics for Medicine Paris, INSERM, ESPCI Paris, PSL Research University, CNRS, France
| |
Collapse
|
9
|
Gudkov SV, Burmistrov DE, Kondakova EV, Sarimov RM, Yarkov RS, Franceschi C, Vedunova MV. An emerging role of astrocytes in aging/neuroinflammation and gut-brain axis with consequences on sleep and sleep disorders. Ageing Res Rev 2023; 83:101775. [PMID: 36334910 DOI: 10.1016/j.arr.2022.101775] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/05/2022] [Accepted: 10/30/2022] [Indexed: 11/18/2022]
Abstract
Understanding the role of astrocytes in the central nervous system has changed dramatically over the last decade. The accumulating findings indicate that glial cells are involved not only in the maintenance of metabolic and ionic homeostasis and in the implementation of trophic functions but also in cognitive functions and information processing in the brain. Currently, there are some controversies regarding the role of astrocytes in complex processes such as aging of the nervous system and the pathogenesis of age-related neurodegenerative diseases. Many findings confirm the important functional role of astrocytes in age-related brain changes, including sleep disturbance and the development of neurodegenerative diseases and particularly Alzheimer's disease. Until recent years, neurobiological research has focused mainly on neuron-glial interactions, in which individual astrocytes locally modulate neuronal activity and communication between neurons. The review considers the role of astrocytes in the physiology of sleep and as an important "player" in the development of neurodegenerative diseases. In addition, the features of the astrocytic network reorganization during aging are discussed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Dmitriy E Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Elena V Kondakova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Ruslan M Sarimov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Roman S Yarkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Maria V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| |
Collapse
|
10
|
Gibson M, Yiallourou S, Pase MP. The Association Between 24-Hour Blood Pressure Profiles and Dementia. J Alzheimers Dis 2023; 94:1303-1322. [PMID: 37458039 DOI: 10.3233/jad-230400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Midlife hypertension increases risk for dementia. Around one third of adults have diagnosed hypertension; however, many adults are undiagnosed, or remain hypertensive despite diagnosis or treatment. Since blood pressure (BP) follows a circadian rhythm, ambulatory BP monitoring allows for the assessment of BP over a 24-hour period and provides an important tool for improving the diagnosis and management of hypertension. The measurement of 24-hour BP profiles, especially nocturnal BP, demonstrate better predictive ability for cardiovascular disease and mortality than office measurement. However, few studies have examined 24-hour BP profiles with respect to dementia risk. This is an important topic since improvements in BP management could facilitate the primary prevention of vascular cognitive impairment and dementia. Therefore, this review discusses the evidence linking BP to dementia, with a focus on whether the implementation of 24-hour BP measurements can improve risk prediction and prevention strategies. Pathways linking nocturnal BP to dementia are also discussed as are risk reduction strategies. Overall, limited research suggests an association between 24-hour BP elevation and poorer cognition, cerebral small vessel disease, and dementia. However, most studies were cross-sectional. Further evidence is needed to substantiate 24-hour BP profiles, over and above office BP, as predictors of vascular cognitive impairment and incident dementia.
Collapse
Affiliation(s)
- Madeline Gibson
- The Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Stephanie Yiallourou
- The Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Matthew P Pase
- The Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
11
|
Venkataraman AV, Mansur A, Rizzo G, Bishop C, Lewis Y, Kocagoncu E, Lingford-Hughes A, Huiban M, Passchier J, Rowe JB, Tsukada H, Brooks DJ, Martarello L, Comley RA, Chen L, Schwarz AJ, Hargreaves R, Gunn RN, Rabiner EA, Matthews PM. Widespread cell stress and mitochondrial dysfunction occur in patients with early Alzheimer's disease. Sci Transl Med 2022; 14:eabk1051. [PMID: 35976998 DOI: 10.1126/scitranslmed.abk1051] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cell stress and impaired oxidative phosphorylation are central to mechanisms of synaptic loss and neurodegeneration in the cellular pathology of Alzheimer's disease (AD). In this study, we quantified the in vivo expression of the endoplasmic reticulum stress marker, sigma 1 receptor (S1R), using [11C]SA4503 positron emission tomography (PET), the mitochondrial complex I (MC1) with [18F]BCPP-EF, and the presynaptic vesicular protein SV2A with [11C]UCB-J in 12 patients with early AD and in 16 cognitively normal controls. We integrated these molecular measures with assessments of regional brain volumes and cerebral blood flow (CBF) measured with magnetic resonance imaging arterial spin labeling. Eight patients with AD were followed longitudinally to estimate the rate of change of the physiological and structural pathology markers with disease progression. The patients showed widespread increases in S1R (≤ 27%) and regional reduction in MC1 (≥ -28%) and SV2A (≥ -25%) radioligand binding, brain volume (≥ -23%), and CBF (≥ -26%). [18F]BCPP-EF PET MC1 binding (≥ -12%) and brain volumes (≥ -5%) showed progressive reductions over 12 to 18 months, suggesting that they both could be used as pharmacodynamic indicators in early-stage therapeutics trials. Associations of reduced MC1 and SV2A and increased S1R radioligand binding with reduced cognitive performance in AD, although exploratory, suggested a loss of metabolic functional reserve with disease. Our study thus provides in vivo evidence for widespread, clinically relevant cellular stress and bioenergetic abnormalities in early AD.
Collapse
Affiliation(s)
- Ashwin V Venkataraman
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK Dementia Research Institute at Imperial College London, London W12 0NN, UK
| | | | - Gaia Rizzo
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,Invicro LLC, London W12 0NN, UK
| | | | | | | | | | | | | | | | - Hideo Tsukada
- Hamamatsu Photonics, Hamakita, Hamamatsu, Shizuoka 4348601, Japan
| | - David J Brooks
- University of Newcastle upon Tyne, Newcastle NE2 4HH, UK.,Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | | | | | | | | | | | - Roger N Gunn
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,Invicro LLC, London W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro LLC, London W12 0NN, UK.,King's College London, London SE5 8AF, UK
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK.,UK Dementia Research Institute at Imperial College London, London W12 0NN, UK
| |
Collapse
|
12
|
Yoo HS, Shanmugalingam U, Smith PD. Potential roles of branched-chain amino acids in neurodegeneration. Nutrition 2022; 103-104:111762. [DOI: 10.1016/j.nut.2022.111762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/12/2022] [Accepted: 05/31/2022] [Indexed: 10/31/2022]
|
13
|
Ni S, Zhang H, Sun L, Zhao Y, Pei C, Nie Y, Liu X, Wu L, Xu A. Transgenerational reproductive toxicity of 2,4,6-trinitrotoluene (TNT) and its metabolite 4-ADNT in Caenorhabditis elegans. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 92:103865. [PMID: 35436606 DOI: 10.1016/j.etap.2022.103865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 06/14/2023]
Abstract
2,4,6-trinitrotoluene (TNT) as an energetic compound widely used in military applications has aroused great concerns in recent years due to its large-scale contamination in soil and water; however, its toxicity is still largely unknown. In this study, we investigated the reproductive toxicity and the transgenerational effects of TNT on Caenorhabditis elegans (C. elegans). Our data showed that exposure to TNT at concentrations ranging from 10 to 100 ng/mL resulted in decreasing the lifespan, brood size, number of oocytes and eggs in uterus, while increasing the number of germ cell apoptosis in C. elegans. The apoptotic effects of TNT were blocked in mutants of cep-1 (w40), egl-1 (n487), and hus-1 (op241), indicating conserved genotoxic response genes was involved in mediating TNT-induced germ cell apoptosis. Parental exposure to TNT significantly increased the germ cell apoptosis from P0 to F2 generation, but the toxicity faded away in F3 and F4 generations. Furthermore, TNT was rapidly metabolized in P0, and the accumulation of 4-aminodinitrotoluene (4-ADNT), the main metabolite of TNT in C. elegans, showed a significant decrease from P0 to F1 and a slow decrease in the subsequent generations. Our results demonstrated that ingested TNT can cause severe transgenerational reproductive toxicity and be rapidly converted to 4-ADNT in the nematodes. These data provided basis for future studies on the effects of energetic compounds across generations.
Collapse
Affiliation(s)
- Shenyao Ni
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, PR China
| | - Huijun Zhang
- Institute of Polar Environment & Anhui Key Laboratory of Polar Environment and Global Change, School of Earth and Space Sciences, University of Science and Technology of China, Hefei, Anhui 230026, PR China
| | - Lingyan Sun
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, PR China
| | - Yanan Zhao
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, PR China
| | - Chengcheng Pei
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, PR China
| | - Yaguang Nie
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, PR China.
| | - Xiaodong Liu
- Institute of Polar Environment & Anhui Key Laboratory of Polar Environment and Global Change, School of Earth and Space Sciences, University of Science and Technology of China, Hefei, Anhui 230026, PR China
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, PR China
| | - An Xu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, PR China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, PR China.
| |
Collapse
|
14
|
Yang M, Sun D, Wang Y, Yan M, Zheng J, Ren J. Cognitive Impairment in Heart Failure: Landscape, Challenges, and Future Directions. Front Cardiovasc Med 2022; 8:831734. [PMID: 35198608 PMCID: PMC8858826 DOI: 10.3389/fcvm.2021.831734] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 12/30/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure (HF) is a major global healthcare problem accounting for substantial deterioration of prognosis. As a complex clinical syndrome, HF often coexists with multi-comorbidities of which cognitive impairment (CI) is particularly important. CI is increasing in prevalence among patients with HF and is present in around 40%, even up to 60%, of elderly patients with HF. As a potent and independent prognostic factor, CI significantly increases the hospitalization and mortality and decreases quality of life in patients with HF. There has been a growing awareness of the complex bidirectional interaction between HF and CI as it shares a number of common pathophysiological pathways including reduced cerebral blood flow, inflammation, and neurohumoral activations. Research that focus on the precise mechanism for CI in HF is still ever insufficient. As the tremendous adverse consequences of CI in HF, effective early diagnosis of CI in HF and interventions for these patients may halt disease progression and improve prognosis. The current clinical guidelines in HF have begun to emphasize the importance of CI. However, nearly half of CI in HF is underdiagnosed, and few recommendations are available to guide clinicians about how to approach CI in patients with HF. This review aims to synthesize knowledge about the link between HF and cognitive dysfunction, issues pertaining to screening, diagnosis and management of CI in patients with HF, and emerging therapies for prevention. Based on data from current studies, critical gaps in knowledge of CI in HF are identified, and future research directions to guide the field forward are proposed.
Collapse
Affiliation(s)
- Mengxi Yang
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Di Sun
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yu Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Mengwen Yan
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jingang Zheng
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jingyi Ren
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
- Vascular Health Research Center of Peking University Health Science Center, Beijing, China
- *Correspondence: Jingyi Ren
| |
Collapse
|
15
|
Borek-Dorosz A, Pieczara A, Czamara K, Stojak M, Matuszyk E, Majzner K, Brzozowski K, Bresci A, Polli D, Baranska M. What is the ability of inflamed endothelium to uptake exogenous saturated fatty acids? A proof-of-concept study using spontaneous Raman, SRS and CARS microscopy. Cell Mol Life Sci 2022; 79:593. [PMID: 36380212 PMCID: PMC9666316 DOI: 10.1007/s00018-022-04616-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/16/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022]
Abstract
Endothelial cells (EC) in vivo buffer and regulate the transfer of plasma fatty acid (FA) to the underlying tissues. We hypothesize that inflammation could alter the functionality of the EC, i.e., their capacity and uptake of different FA. The aim of this work is to verify the functionality of inflamed cells by analyzing their ability to uptake and accumulate exogenous saturated FA. Control and inflammatory human microvascular endothelial cells stimulated in vitro with two deuterium-labeled saturated FA (D-FA), i.e., palmitic (D31-PA) and myristic (D27-MA) acids. Cells were measured both by spontaneous and stimulated Raman imaging to extract detailed information about uptaken FA, whereas coherent anti-Stokes Raman scattering and fluorescence imaging showed the global content of FA in cells. Additionally, we employed atomic force microscopy to obtain a morphological image of the cells. The results indicate that the uptake of D-FA in inflamed cells is dependent on their concentration and type. Cells accumulated D-FA when treated with a low concentration, and the effect was more pronounced for D27-MA, in normal cells, but even more so, in inflamed cells. In the case of D31-PA, a slightly increased uptake was observed for inflamed cells when administered at higher concentration. The results provide a better understanding of the EC inflammation and indicate the impact of the pathological state of the EC on their capacity to buffer fat. All the microscopic methods used showed complementarity in the analysis of FA uptake by EC, but each method recognized this process from a different perspective.
Collapse
Affiliation(s)
| | - Anna Pieczara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Krzysztof Czamara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Ewelina Matuszyk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Katarzyna Majzner
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland ,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| | - Krzysztof Brzozowski
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland
| | - Arianna Bresci
- Physics Department, Politecnico di Milano, Piazza Leonardo da Vinci, 32, 20133 Milan, Italy
| | - Dario Polli
- Physics Department, Politecnico di Milano, Piazza Leonardo da Vinci, 32, 20133 Milan, Italy ,Institute for Photonics and Nanotechnology at CNR (CNR-IFN), Piazza Leonardo da Vinci, 32, 20133 Milan, Italy
| | - Malgorzata Baranska
- Faculty of Chemistry, Jagiellonian University, 2 Gronostajowa Str., 30-387 Krakow, Poland ,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego Str., 30-348 Krakow, Poland
| |
Collapse
|
16
|
Pellegrini C, D’Antongiovanni V, Fornai M, Duranti E, Baldacci F, Bernardini N, Taddei S, Virdis A, Blandizzi C, Masi S, Antonioli L. Donepezil improves vascular function in a mouse model of Alzheimer's disease. Pharmacol Res Perspect 2021; 9:e00871. [PMID: 34713597 PMCID: PMC8554410 DOI: 10.1002/prp2.871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/14/2021] [Indexed: 01/29/2023] Open
Abstract
Cardiovascular complications in Alzheimer's disease (AD) patients can occur years to decades prior to the onset of clinical symptoms of the disease. Donepezil represents the most effective drug in the treatment of AD. However, the potential effect of donepezil on vascular function and structure remains largely unexplored. Here, we assessed the impact of donepezil on the vascular phenotype of an established model of accelerated senescence that develops spontaneously AD, the SAMP8 mouse. Three groups of animals were included: SAMR1 (control strain), SAMP8, and SAMP8 treated with donepezil. Treatment with donepezil was administered from the 4th to the 6th month of life. At 6 months, after cognitive tests by Morris Water Maze, animals were euthanized, and their mesenteric arteries were processed for functional experiments. Untreated SAMP8 developed cognitive impairment compared to SAMR1, while donepezil treatment significantly attenuated cognitive dysfunction. SAMP8 exhibited a higher media-to-lumen ratio than SAMR1 and donepezil-treated animals. Endothelial function was impaired in SAMP8 animals compared to SAMR1. The addition of vitamin C improved the vasodilatory response to acetylcholine in SAMP8. Treatment with donepezil improved endothelial function in SAMP8 animals and reduced the additional vasodilation induced by vitamin C. In conclusion, in the SAMP8 AD model, cognitive impairment is associated with endothelial dysfunction and vascular remodeling which could contribute to cardiovascular events in AD since the prodromal phases of the disease. Treatment with donepezil alleviates vascular dysfunction associated with AD through an increase in NO availability likely by counteracting inflammation and oxidative stress.
Collapse
Affiliation(s)
- Carolina Pellegrini
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| | - Vanessa D’Antongiovanni
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| | - Matteo Fornai
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| | - Emiliano Duranti
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| | - Filippo Baldacci
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| | - Nunzia Bernardini
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
- Interdepartmental Research Center “Nutraceuticals and Food for Health”University of PisaPisaItaly
| | - Stefano Taddei
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| | - Agostino Virdis
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| | - Corrado Blandizzi
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| | - Stefano Masi
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| | - Luca Antonioli
- Department of Clinical and Experimental MedicineSchool of MedicineUniversity of PisaPisaItaly
| |
Collapse
|
17
|
Vittal Rao H, Bihaqi SW, Iannucci J, Sen A, Grammas P. Thrombin Signaling Contributes to High Glucose-Induced Injury of Human Brain Microvascular Endothelial Cells. J Alzheimers Dis 2021; 79:211-224. [PMID: 33252072 DOI: 10.3233/jad-200658] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Diabetes is one of the strongest disease-related risk factors for Alzheimer's disease (AD). In diabetics, hyperglycemia-induced microvascular complications are the major cause of end-organ injury, contributing to morbidity and mortality. Microvascular pathology is also an important and early feature of AD. The cerebral microvasculature may be a point of convergence of both diseases. Several lines of evidence also implicate thrombin in AD as well as in diabetes. OBJECTIVE Our objective was to investigate the role of thrombin in glucose-induced brain microvascular endothelial injury. METHODS Cultured Human brain microvascular endothelial cells (HBMVECs) were treated with 30 mM glucose±100 nM thrombin and±250 nM Dabigatran or inhibitors of PAR1, p38MAPK, MMP2, or MMP9. Cytotoxicity and thrombin activity assays on supernatants and western blotting for protein expression in lysates were performed. RESULTS reatment of HBMVECs with 30 mM glucose increased thrombin activity and expression of inflammatory proteins TNFα, IL-6, and MMPs 2 and 9; this elevation was reduced by the thrombin inhibitor dabigatran. Direct treatment of brain endothelial cells with thrombin upregulated p38MAPK and CREB, and induced TNFα, IL6, MMP2, and MMP9 as well as oxidative stress proteins NOX4 and iNOS. Inhibition of thrombin, thrombin receptor PAR1 or p38MAPK decrease expression of inflammatory and oxidative stress proteins, implying that thrombin may play a central role in glucose-induced endothelial injury. CONCLUSION Since preventing brain endothelial injury would preserve blood-brain barrier integrity, prevent neuroinflammation, and retain intact functioning of the neurovascular unit, inhibiting thrombin, or its downstream signaling effectors, could be a therapeutic strategy for mitigating diabetes-induced dementia.
Collapse
Affiliation(s)
- Haripriya Vittal Rao
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston Salem, NC, USA.,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Syed Waseem Bihaqi
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA.,Department of Neuroscience & Regenerative Medicine, Augusta University, Augusta, GA, USA
| | - Jaclyn Iannucci
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA.,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Abhik Sen
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA.,Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Paula Grammas
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA.,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
18
|
Cognitive Dysfunction after Heart Disease: A Manifestation of the Heart-Brain Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4899688. [PMID: 34457113 PMCID: PMC8387198 DOI: 10.1155/2021/4899688] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/31/2021] [Indexed: 12/26/2022]
Abstract
The functions of the brain and heart, which are the two main supporting organs of human life, are closely linked. Numerous studies have expounded the mechanisms of the brain-heart axis and its related clinical applications. However, the effect of heart disease on brain function, defined as the heart-brain axis, is less studied even though cognitive dysfunction after heart disease is one of its most frequently reported manifestations. Hypoperfusion caused by heart failure appears to be an important risk factor for cognitive decline. Blood perfusion, the immune response, and oxidative stress are the possible main mechanisms of cognitive dysfunction, indicating that the blood-brain barrier, glial cells, and amyloid-β may play active roles in these mechanisms. Clinicians should pay more attention to the cognitive function of patients with heart disease, especially those with heart failure. In addition, further research elucidating the associated mechanisms would help discover new therapeutic targets to intervene in the process of cognitive dysfunction after heart disease. This review discusses cognitive dysfunction in relation to heart disease and its potential mechanisms.
Collapse
|
19
|
Huang P, He XY, Xu M. Dengzhan shengmai capsule combined with donepezil hydrochloride in the treatment of Alzheimer's disease: preliminary findings, randomized and controlled clinical trial. ACTA ACUST UNITED AC 2021; 67:190-194. [PMID: 34287475 DOI: 10.1590/1806-9282.67.02.20200378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 11/16/2020] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To observe the effects of Dengzhan Shengmai capsule combined with donepezil hydrochloride on cognitive function, daily living ability, and safety in patients with Alzheimer's disease. METHODS A total of 294 patients with Alzheimer's disease were randomly divided into a treatment group and a control group, 147 cases each group. The control group was given oral donepezil hydrochloride 5 mg once a day, and the treatment group was given oral Dengzhan Shengmai capsule 0.36 g three times a day, based on the control group. RESULTS At 3 and 6 months of treatment, the ADAS-cog score of the treatment group was 48.69±6.23 and 44.24±5.53; for the control group, 45.48±5.94 and 41.57±5.10. The difference between the two groups is statistically significant (p<0.05). At 3 and 6 months of treatment, the NO level in the treatment group was (46.28±6.68) umol/l, (43.55±7.92) umol/l, and the control group was (42.95±7.92) umol/l, (38.89±5.93) umol/l. The differences between both groups were statistically significant (p<0.05). At 3 and 6 months of treatment, ET levels in the treatment group were (156.08±17.39) ng/l, (144.91±17.60) ng/l, and the control group was (150.48±22.94) ng/l, (135.04±10.08) ng/l. Correlation analysis showed that ADAS-cog score was negatively correlated with NO and ET (p<0.001). CONCLUSIONS Dengzhan Shengmai capsule combined with donepezil hydrochloride can improve cognitive function and the living capacity of patients with Alzheimer's disease, reduce the production of neurotoxic substances NO and ET, and provide higher safety.
Collapse
Affiliation(s)
- Pan Huang
- People1s Hospital of DeYang City, Department of Neurology - DeYang, China
| | - Xiao-Ying He
- The Affiliated Hospital of Southwest Medical University, Department of Neurology - Luzhou, China
| | - Min Xu
- The second People's Hospital of DeYang City, Department of Neurology - DeYang, China
| |
Collapse
|
20
|
Sharma HS, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Buzoianu AD, Sahib S, Tian ZR, Bryukhovetskiy I, Manzhulo I, Menon PK, Patnaik R, Wiklund L, Sharma A. Alzheimer's disease neuropathology is exacerbated following traumatic brain injury. Neuroprotection by co-administration of nanowired mesenchymal stem cells and cerebrolysin with monoclonal antibodies to amyloid beta peptide. PROGRESS IN BRAIN RESEARCH 2021; 265:1-97. [PMID: 34560919 DOI: 10.1016/bs.pbr.2021.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Military personnel are prone to traumatic brain injury (TBI) that is one of the risk factors in developing Alzheimer's disease (AD) at a later stage. TBI induces breakdown of the blood-brain barrier (BBB) to serum proteins into the brain and leads to extravasation of plasma amyloid beta peptide (ΑβP) into the brain fluid compartments causing AD brain pathology. Thus, there is a need to expand our knowledge on the role of TBI in AD. In addition, exploration of the novel roles of nanomedicine in AD and TBI for neuroprotection is the need of the hour. Since stem cells and neurotrophic factors play important roles in TBI and in AD, it is likely that nanodelivery of these agents exert superior neuroprotection in TBI induced exacerbation of AD brain pathology. In this review, these aspects are examined in details based on our own investigations in the light of current scientific literature in the field. Our observations show that TBI exacerbates AD brain pathology and TiO2 nanowired delivery of mesenchymal stem cells together with cerebrolysin-a balanced composition of several neurotrophic factors and active peptide fragments, and monoclonal antibodies to amyloid beta protein thwarted the development of neuropathology following TBI in AD, not reported earlier.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
21
|
Luo Z, Xu H, Liu L, Ohulchanskyy TY, Qu J. Optical Imaging of Beta-Amyloid Plaques in Alzheimer's Disease. BIOSENSORS 2021; 11:255. [PMID: 34436057 PMCID: PMC8392287 DOI: 10.3390/bios11080255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 02/02/2023]
Abstract
Alzheimer's disease (AD) is a multifactorial, irreversible, and incurable neurodegenerative disease. The main pathological feature of AD is the deposition of misfolded β-amyloid protein (Aβ) plaques in the brain. The abnormal accumulation of Aβ plaques leads to the loss of some neuron functions, further causing the neuron entanglement and the corresponding functional damage, which has a great impact on memory and cognitive functions. Hence, studying the accumulation mechanism of Aβ in the brain and its effect on other tissues is of great significance for the early diagnosis of AD. The current clinical studies of Aβ accumulation mainly rely on medical imaging techniques, which have some deficiencies in sensitivity and specificity. Optical imaging has recently become a research hotspot in the medical field and clinical applications, manifesting noninvasiveness, high sensitivity, absence of ionizing radiation, high contrast, and spatial resolution. Moreover, it is now emerging as a promising tool for the diagnosis and study of Aβ buildup. This review focuses on the application of the optical imaging technique for the determination of Aβ plaques in AD research. In addition, recent advances and key operational applications are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Junle Qu
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China; (Z.L.); (H.X.); (L.L.); (T.Y.O.)
| |
Collapse
|
22
|
Chemical Composition, In Vitro and In Silico Antioxidant Potential of Melissa officinalis subsp. officinalis Essential Oil. Antioxidants (Basel) 2021; 10:antiox10071081. [PMID: 34356313 PMCID: PMC8301138 DOI: 10.3390/antiox10071081] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 01/16/2023] Open
Abstract
The investigation aimed to study the in vitro and in silico antioxidant properties of Melissa officinalis subsp. officinalis essential oil (MOEO). The chemical composition of MOEO was determined using GC–MS analysis. Among 36 compounds identified in MOEO, the main were beta-cubebene (27.66%), beta-caryophyllene (27.41%), alpha-cadinene (4.72%), caryophyllene oxide (4.09%), and alpha-cadinol (4.07%), respectively. In vitro antioxidant properties of MOEO have been studied in 2,2’-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and 2,2-diphenyl-1-picrylhydrazyl (DPPH) free-radical scavenging, and inhibition of β-carotene bleaching assays. The half-maximal inhibitory concentration (IC50) for the radical scavenging abilities of ABTS and DPPH were 1.225 ± 0.011 μg/mL and 14.015 ± 0.027 μg/mL, respectively, demonstrating good antioxidant activity. Moreover, MOEO exhibited a strong inhibitory effect (94.031 ± 0.082%) in the β-carotene bleaching assay by neutralizing hydroperoxides, responsible for the oxidation of highly unsaturated β-carotene. Furthermore, molecular docking showed that the MOEO components could exert an in vitro antioxidant activity through xanthine oxidoreductase inhibition. The most active structures are minor MOEO components (approximately 6%), among which the highest affinity for the target protein belongs to carvacrol.
Collapse
|
23
|
The Microbiota-Gut-Brain Axis and Alzheimer Disease. From Dysbiosis to Neurodegeneration: Focus on the Central Nervous System Glial Cells. J Clin Med 2021; 10:jcm10112358. [PMID: 34072107 PMCID: PMC8199461 DOI: 10.3390/jcm10112358] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
The microbiota-gut system can be thought of as a single unit that interacts with the brain via the "two-way" microbiota-gut-brain axis. Through this axis, a constant interplay mediated by the several products originating from the microbiota guarantees the physiological development and shaping of the gut and the brain. In the present review will be described the modalities through which the microbiota and gut control each other, and the main microbiota products conditioning both local and brain homeostasis. Much evidence has accumulated over the past decade in favor of a significant association between dysbiosis, neuroinflammation and neurodegeneration. Presently, the pathogenetic mechanisms triggered by molecules produced by the altered microbiota, also responsible for the onset and evolution of Alzheimer disease, will be described. Our attention will be focused on the role of astrocytes and microglia. Numerous studies have progressively demonstrated how these glial cells are important to ensure an adequate environment for neuronal activity in healthy conditions. Furthermore, it is becoming evident how both cell types can mediate the onset of neuroinflammation and lead to neurodegeneration when subjected to pathological stimuli. Based on this information, the role of the major microbiota products in shifting the activation profiles of astrocytes and microglia from a healthy to a diseased state will be discussed, focusing on Alzheimer disease pathogenesis.
Collapse
|
24
|
Chubarev VN, Beeraka NM, Sinelnikov MY, Bulygin KV, Nikolenko VN, Mihaylenko E, Tarasov VV, Mikhaleva LM, Poltronieri P, Viswanadha VP, Somasundaram SG, Kirkland CE, Chen K, Liu J, Fan R, Kamal MA, Mironov AA, Madhunapantula SV, Pretorius E, Dindyaev SV, Muresanu C, Sukocheva OA. Health Science Community Will Miss This Bright and Uniting Star: In Memory of Professor Gjumrakch Aliev, M.D, Ph.D. Cancers (Basel) 2021; 13:cancers13081965. [PMID: 33921833 PMCID: PMC8072812 DOI: 10.3390/cancers13081965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 02/05/2023] Open
Abstract
It is with deep sadness that we offer our memorial on the unexpected demise of our dear colleague, Professor Gjumrakch Aliev [...].
Collapse
Affiliation(s)
- Vladimir N. Chubarev
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | - Narasimha M. Beeraka
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Bannimantapa, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka 570 015, India;
| | - Mikhail Y. Sinelnikov
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | - Kirill V. Bulygin
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
- Faculty of Medicine, M.V. Lomonosov Moscow State University, 117192 Moscow, Russia
| | - Vladimir N. Nikolenko
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
- Faculty of Medicine, M.V. Lomonosov Moscow State University, 117192 Moscow, Russia
| | - Elizaveta Mihaylenko
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | - Vadim V. Tarasov
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | | | - Palmiro Poltronieri
- Institute of Sciences of Food Productions, National Research Council of Italy, via Monteroni km 7, 73100 Lecce, Italy;
| | | | - Siva G. Somasundaram
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Cecil E. Kirkland
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Kuo Chen
- Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; (K.C.); (J.L.); (R.F.)
| | - Junqi Liu
- Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; (K.C.); (J.L.); (R.F.)
| | - Ruitai Fan
- Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; (K.C.); (J.L.); (R.F.)
| | - Mohammad Amjad Kamal
- West China School of Nursing/Institutes for Systems Genetics, The Frontier Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China;
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- Enzymoics, 7 Peterlee Place, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Alexander A. Mironov
- Laboratory of Electron Microscopy, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy;
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Bannimantapa, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka 570 015, India;
| | - Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa;
| | - Sergey V. Dindyaev
- Department of Histology, Embryology & Cytology, Pediatric Faculty, Federal State Budgetary Educational Institution of Higher Education “Ivanovo State Medical Academy” of the Ministry of Healthcare of the Russian Federation (FSBEI HE IvSMA MOH Russia), 8 Sheremetyevsky Ave., 153012 Ivanovo, Russia;
| | - Cristian Muresanu
- Research Center for Applied Biotechnology in Diagnosis and Molecular Therapies, Str. Trifoiului nr. 12 G, 400478 Cluj-Napoca, Romania;
| | - Olga A. Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University of South Australia, Adelaide 5001, Australia
- Correspondence:
| |
Collapse
|
25
|
Lana D, Ugolini F, Giovannini MG. An Overview on the Differential Interplay Among Neurons-Astrocytes-Microglia in CA1 and CA3 Hippocampus in Hypoxia/Ischemia. Front Cell Neurosci 2020; 14:585833. [PMID: 33262692 PMCID: PMC7686560 DOI: 10.3389/fncel.2020.585833] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons have been long regarded as the basic functional cells of the brain, whereas astrocytes and microglia have been regarded only as elements of support. However, proper intercommunication among neurons-astrocytes-microglia is of fundamental importance for the functional organization of the brain. Perturbation in the regulation of brain energy metabolism not only in neurons but also in astrocytes and microglia may be one of the pathophysiological mechanisms of neurodegeneration, especially in hypoxia/ischemia. Glial activation has long been considered detrimental for survival of neurons, but recently it appears that glial responses to an insult are not equal but vary in different brain areas. In this review, we first take into consideration the modifications of the vascular unit of the glymphatic system and glial metabolism in hypoxic conditions. Using the method of triple-labeling fluorescent immunohistochemistry coupled with confocal microscopy (TIC), we recently studied the interplay among neurons, astrocytes, and microglia in chronic brain hypoperfusion. We evaluated the quantitative and morpho-functional alterations of the neuron-astrocyte-microglia triads comparing the hippocampal CA1 area, more vulnerable to ischemia, to the CA3 area, less vulnerable. In these contiguous and interconnected areas, in the same experimental hypoxic conditions, astrocytes and microglia show differential, finely regulated, region-specific reactivities. In both areas, astrocytes and microglia form triad clusters with apoptotic, degenerating neurons. In the neuron-astrocyte-microglia triads, the cell body of a damaged neuron is infiltrated and bisected by branches of astrocyte that create a microscar around it while a microglial cell phagocytoses the damaged neuron. These coordinated actions are consistent with the scavenging and protective activities of microglia. In hypoxia, the neuron-astrocyte-microglia triads are more numerous in CA3 than in CA1, further indicating their protective effects. These data, taken from contiguous and interconnected hippocampal areas, demonstrate that glial response to the same hypoxic insult is not equal but varies significantly. Understanding the differences of glial reactivity is of great interest to explain the differential susceptibility of hippocampal areas to hypoxia/ischemia. Further studies may evidence the differential reactivity of glia in different brain areas, explaining the higher or lower sensitivity of these areas to different insults and whether glia may represent a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Florence, Italy
| | - Maria G Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
26
|
Tobore TO. On the Etiopathogenesis and Pathophysiology of Alzheimer's Disease: A Comprehensive Theoretical Review. J Alzheimers Dis 2020; 68:417-437. [PMID: 30775973 DOI: 10.3233/jad-181052] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alzheimers' disease (AD) is the most common cause of dementia, with an estimated 5 million new cases occurring annually. Among the elderly, AD shortens life expectancy, results in disability, decreases quality of life, and ultimately, leads to institutionalization. Despite extensive research in the last few decades, its heterogeneous pathophysiology and etiopathogenesis have made it difficult to develop an effective treatment and prevention strategy. Aging is the biggest risk factor for AD and evidence suggest that the total number of older people in the population is going to increase astronomically in the next decades. Also, there is evidence that air pollution and increasing income inequality may result in higher incidence and prevalence of AD. This makes the need for a comprehensive understanding of the etiopathogenesis and pathophysiology of the disease extremely critical. In this paper, a quintuple framework of thyroid dysfunction, vitamin D deficiency, sex hormones, and mitochondria dysfunction and oxidative stress are used to provide a comprehensive description of AD etiopathogenesis and pathophysiology. The individual role of each factor, their synergistic and genetic interactions, as well as the limitations of the framework are discussed.
Collapse
|
27
|
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: 60] [Impact Index Per Article: 15.0] [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
|
28
|
Abstract
PURPOSE OF REVIEW This review summarizes the evidence for the established vascular/hypoperfusion model and explores the new hypothesis that configures the heart/brain axis as an organ system where similar pathogenic mechanisms exploit physiological and pathological changes. RECENT FINDINGS Although associated by common risk factors, similar epidemiological stratification and common triggers (including inflammation, oxidative stress, and hypoxia), heart failure and Alzheimer's disease have been, for long time, viewed as pathogenically separate illnesses. The silos began to be broken down with the awareness that vascular dysfunction, and loss of cardiac perfusion pump power, trigger biochemical changes, contributing to the typical hallmark of Alzheimer's disease (AD)-the accumulation of Aβ plaques and hyperphosphorylated Tau tangles. Compromised blood flow to the brain becomes the paradigm for the "heart-to-head" connection. Compelling evidence of common genetic variants, biochemical characteristics, and the accumulation of Aβ outside the brain suggests a common pathogenesis for heart failure (HF) and AD. These new findings represent just the beginning of the understanding the complex connection between AD and HF requiring further studies and interdisciplinary approaches. Altogether, the current evidence briefly summarized in this review, highlight a closer and complex relationship between heart failure and Alzheimer's that goes beyond the vascular/perfusion hypothesis. Genetic and biochemical evidence begin to suggest common pathogenic mechanisms between the two diseases involving a systemic defect in the folding of protein or a seeding at distance of the misfolded proteins from one organ to the other.
Collapse
|
29
|
Kodavati M, Wang H, Hegde ML. Altered Mitochondrial Dynamics in Motor Neuron Disease: An Emerging Perspective. Cells 2020; 9:cells9041065. [PMID: 32344665 PMCID: PMC7226538 DOI: 10.3390/cells9041065] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondria plays privotal role in diverse pathways that regulate cellular function and survival, and have emerged as a prime focus in aging and age-associated motor neuron diseases (MNDs), such as amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Accumulating evidence suggests that many amyloidogenic proteins, including MND-associated RNA/DNA-binding proteins fused in sarcoma (FUS) and TAR DNA binding protein (TDP)-43, are strongly linked to mitochondrial dysfunction. Animal model and patient studies have highlighted changes in mitochondrial structure, plasticity, replication/copy number, mitochondrial DNA instability, and altered membrane potential in several subsets of MNDs, and these observations are consistent with the evidence of increased excitotoxicity, induction of reactive oxygen species, and activation of intrinsic apoptotic pathways. Studies in MND rodent models also indicate that mitochondrial abnormalities begin prior to the clinical and pathological onset of the disease, suggesting a causal role of mitochondrial dysfunction. Our recent studies, which demonstrated the involvement of specific defects in DNA break-ligation mediated by DNA ligase 3 (LIG3) in FUS-associated ALS, raised a key question of its potential implication in mitochondrial DNA transactions because LIG3 is essential for both mitochondrial DNA replication and repair. This question, as well as how wild-type and mutant MND-associated factors affect mitochondria, remain to be elucidated. These new investigation avenues into the mechanistic role of mitochondrial dysfunction in MNDs are critical to identify therapeutic targets to alleviate mitochondrial toxicity and its consequences. In this article, we critically review recent advances in our understanding of mitochondrial dysfunction in diverse subgroups of MNDs and discuss challenges and future directions.
Collapse
Affiliation(s)
- Manohar Kodavati
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (M.K.); (H.W.)
| | - Haibo Wang
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (M.K.); (H.W.)
| | - Muralidhar L. Hegde
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (M.K.); (H.W.)
- Department of Neurosurgery, Weill Medical College, New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
30
|
Muddapu VR, Dharshini SAP, Chakravarthy VS, Gromiha MM. Neurodegenerative Diseases - Is Metabolic Deficiency the Root Cause? Front Neurosci 2020; 14:213. [PMID: 32296300 PMCID: PMC7137637 DOI: 10.3389/fnins.2020.00213] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/26/2020] [Indexed: 01/31/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer, Parkinson, Huntington, and amyotrophic lateral sclerosis, are a prominent class of neurological diseases currently without a cure. They are characterized by an inexorable loss of a specific type of neurons. The selective vulnerability of specific neuronal clusters (typically a subcortical cluster) in the early stages, followed by the spread of the disease to higher cortical areas, is a typical pattern of disease progression. Neurodegenerative diseases share a range of molecular and cellular pathologies, including protein aggregation, mitochondrial dysfunction, glutamate toxicity, calcium load, proteolytic stress, oxidative stress, neuroinflammation, and aging, which contribute to neuronal death. Efforts to treat these diseases are often limited by the fact that they tend to address any one of the above pathological changes while ignoring others. Lack of clarity regarding a possible root cause that underlies all the above pathologies poses a significant challenge. In search of an integrative theory for neurodegenerative pathology, we hypothesize that metabolic deficiency in certain vulnerable neuronal clusters is the common underlying thread that links many dimensions of the disease. The current review aims to present an outline of such an integrative theory. We present a new perspective of neurodegenerative diseases as metabolic disorders at molecular, cellular, and systems levels. This helps to understand a common underlying mechanism of the many facets of the disease and may lead to more promising disease-modifying therapeutic interventions. Here, we briefly discuss the selective metabolic vulnerability of specific neuronal clusters and also the involvement of glia and vascular dysfunctions. Any failure in satisfaction of the metabolic demand by the neurons triggers a chain of events that precipitate various manifestations of neurodegenerative pathology.
Collapse
Affiliation(s)
- Vignayanandam Ravindernath Muddapu
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - S. Akila Parvathy Dharshini
- Protein Bioinformatics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - V. Srinivasa Chakravarthy
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - M. Michael Gromiha
- Protein Bioinformatics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
31
|
Birla H, Minocha T, Kumar G, Misra A, Singh SK. Role of Oxidative Stress and Metal Toxicity in the Progression of Alzheimer's Disease. Curr Neuropharmacol 2020; 18:552-562. [PMID: 31969104 PMCID: PMC7457422 DOI: 10.2174/1570159x18666200122122512] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/18/2019] [Accepted: 01/14/2020] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is one of the life-threatening neurodegenerative disorders in the elderly (>60 years) and incurable across the globe to date. AD is caused by the involvement of various genetic, environmental and lifestyle factors that affect neuronal cells to degenerate over the period of time. The oxidative stress is engaged in the pathogenesis of various disorders and its key role is also linked to the etiology of AD. AD is attributed by neuronal loss, abnormal accumulation of Amyloid-β (Aβ) and neurofibrillary tangles (NFTs) with severe memory impairments and other cognitive dysfunctions which lead to the loss of synapses and neuronal death and eventual demise of the individual. Increased production of reactive oxygen species (ROS), loss of mitochondrial function, altered metal homeostasis, aberrant accumulation of senile plaque and mitigated antioxidant defense mechanism all are indulged in the progression of AD. In spite of recent advances in biomedical research, the underlying mechanism of disruption of redox balance and the actual source of oxidative stress is still obscure. This review highlights the generation of ROS through different mechanisms, the role of some important metals in the progression of AD and free radical scavenging by endogenous molecule and supplementation of nutrients in AD.
Collapse
Affiliation(s)
| | | | | | | | - Sandeep Kumar Singh
- Address correspondence to this author at the Indian Scientific Education and Technology Foundation, Lucknow-226002, India;E-mails: ;
| |
Collapse
|
32
|
Shin Y, Choi SH, Kim E, Bylykbashi E, Kim JA, Chung S, Kim DY, Kamm RD, Tanzi RE. Blood-Brain Barrier Dysfunction in a 3D In Vitro Model of Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900962. [PMID: 31637161 PMCID: PMC6794630 DOI: 10.1002/advs.201900962] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/20/2019] [Indexed: 05/21/2023]
Abstract
Harmful materials in the blood are prevented from entering the healthy brain by a highly selective blood-brain barrier (BBB), and impairment of barrier function has been associated with a variety of neurological diseases. In Alzheimer's disease (AD), BBB breakdown has been shown to occur even before cognitive decline and brain pathology. To investigate the role of the cerebral vasculature in AD, a physiologically relevant 3D human neural cell culture microfluidic model is developed having a brain endothelial cell monolayer with a BBB-like phenotype. This model is shown to recapitulate several key aspects of BBB dysfunction observed in AD patients: increased BBB permeability, decreased expression of claudin-1, claudin-5, and VE-cadherin, increased expression of matrix-metalloproteinase-2 and reactive oxygen species, and deposition of β-amyloid (Aβ) peptides at the vascular endothelium. Thus, it provides a well-controlled platform for investigating BBB function as well as for screening of new drugs that need to pass the BBB to gain access to neural tissues.
Collapse
Affiliation(s)
- Yoojin Shin
- Department of Mechanical EngineeringMassachusetts Institute of Technology500 Technology Square, MIT Building, Room NE47‐321CambridgeMA02139USA
| | - Se Hoon Choi
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Eunhee Kim
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Enjana Bylykbashi
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Jeong Ah Kim
- Biomedical Omics GroupKorea Basic Science InstituteCheongju28119Republic of Korea
- Department of Bio‐Analytical ScienceUniversity of Science and TechnologyDaejeon34113Republic of Korea
| | - Seok Chung
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
- School of Mechanical EngineeringKorea UniversitySeoul02841Republic of Korea
| | - Doo Yeon Kim
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| | - Roger D. Kamm
- Department of Mechanical EngineeringMassachusetts Institute of Technology500 Technology Square, MIT Building, Room NE47‐321CambridgeMA02139USA
- Department of Biological EngineeringMassachusetts Institute of Technology500 Technology Square, MIT Building, Room NE47‐321CambridgeMA02139USA
- Singapore‐MIT Alliance for Research & Technology (SMART)BioSystems and Micromechanics (BioSyM)Singapore138602Singapore
| | - Rudolph E. Tanzi
- Genetics and Aging Research UnitMcCance Center for Brain HealthMass General Institute for Neurodegenerative DiseaseDepartment of NeurologyMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02129USA
| |
Collapse
|
33
|
Xiao X, Zhang X, Zhang C, Li J, Zhao Y, Zhu Y, Zhang J, Zhou X. Toxicity and multigenerational effects of bisphenol S exposure to Caenorhabditis elegans on developmental, biochemical, reproductive and oxidative stress. Toxicol Res (Camb) 2019; 8:630-640. [PMID: 31559007 DOI: 10.1039/c9tx00055k] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/21/2019] [Indexed: 12/28/2022] Open
Abstract
Bisphenol A (BPA) is a typical endocrine disruptor. Bisphenol S (BPS) has been widely used as a substitute for various plastic materials due to the limited application of BPA. However, it does not mean that BPS is a safe substitute due to the lack of effective evaluation of BPS. In this study, the clinical model of Caenorhabditis elegans (C. elegans) was used to study the effects of BPS on the locomotion behavior, growth, reproduction, lifespan and antioxidant system. Our study found that C. elegans exposed to 0.01 μM BPS could have significantly inhibited locomotion behavior and growth, as well as damaged reproductive and antioxidant systems and lifespan. It is interesting to note that in multi-generational exposure studies, we found that BPS exhibits complex genotoxicity. With the transmission to the offspring, BPS showed more significant inhibition of the head thrashes of the nematode, while the effect on the body bends and body length was gradually weakened. The effect of BPS on the brood size shows different rules according to different concentrations and offsprings. Therefore, the safety of BPS still needs further evaluation, especially the multi-generational genotoxicity.
Collapse
Affiliation(s)
- Xiang Xiao
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang 212013 , PR China . ; ; Tel: +86-511-88797202
| | - Xiaowei Zhang
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang 212013 , PR China . ; ; Tel: +86-511-88797202
| | - Caiqin Zhang
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang 212013 , PR China . ; ; Tel: +86-511-88797202
| | - Jie Li
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang 212013 , PR China . ; ; Tel: +86-511-88797202
| | - Yansheng Zhao
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang 212013 , PR China . ; ; Tel: +86-511-88797202
| | - Ying Zhu
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang 212013 , PR China . ; ; Tel: +86-511-88797202
| | - Jiayan Zhang
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang 212013 , PR China . ; ; Tel: +86-511-88797202
| | - Xinghua Zhou
- School of Food and Biological Engineering , Jiangsu University , Zhenjiang 212013 , PR China . ; ; Tel: +86-511-88797202
| |
Collapse
|
34
|
Bukiya AN. Fetal Cerebral Artery Mitochondrion as Target of Prenatal Alcohol Exposure. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16091586. [PMID: 31067632 PMCID: PMC6539770 DOI: 10.3390/ijerph16091586] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022]
Abstract
Prenatal alcohol exposure results in an array of developmental abnormalities known as fetal alcohol spectrum disorders (FASDs). Despite the high prevalence of FASDs, therapeutic interventions against accidental or intended exposure of developing fetuses to alcohol are limited. This review outlines current knowledge about mitochondria in cerebral blood vessels as a potential target for anti-FASDs intervention. First, it describes the multifaceted role of mitochondria in maintaining the cerebral artery diameter as shown in adult tissue. Second, current literature on alcohol-driven damage of mitochondrial morphology and function in several fetal tissues, including liver, heart, and brain is summarized. The functional consequences of alcohol exposure in these organs include morphological enlargement of mitochondria, increased oxidative stress, and alteration of cellular respiration. These studies point to a tissue-specific effect of alcohol on mitochondrial function and a particular vulnerability of fetal mitochondria to alcohol exposure when compared to adult counterparts. Third, recent work from our group describing persistent changes in fetal baboon cerebral artery proteome following three episodes of prenatal alcohol exposure is reviewed. In conclusion, the consequences of prenatal alcohol exposure on cerebral artery mitochondria constitute an open field of investigation and, eventually, a point of therapeutic intervention against FASDs.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
35
|
Abstract
In a somewhat narrow diagnostic lens, Alzheimer disease (AD) has been considered a brain-specific disease characterized by the presence of Aβ (β-amyloid) plaques and tau neural fibrillary tangles and neural inflammation; these pathologies lead to neuronal death and consequently clinical symptoms, such as memory loss, confusion, and impaired cognitive function. However, for decades, researchers have noticed a link between various cardiovascular abnormalities and AD-such as heart failure, coronary artery disease, atrial fibrillation, and vasculopathy. A considerable volume of work has pointed at this head to heart connection, focusing mainly on associations between cerebral hypoperfusion and neuronal degradation. However, new evidence of a possible systemic or metastatic profile to AD calls for further analysis of this connection. Aβ aggregations-biochemically and structurally akin to those found in the typical AD pathology-are now known to be present in the hearts of individuals with idiopathic dilated cardiomyopathy, as well as the hearts of patients with AD. These findings suggest a potential systemic profile of proteinopathies and a new hypothesis for the link between peripheral and central symptoms of heart failure and AD. Herein, we provide an overview of the cardiovascular links to Alzheimer disease.
Collapse
Affiliation(s)
- Joshua M Tublin
- From the College of Nursing (J.M.T., J.M.A., L.E.W.), The Ohio State University, Columbus
| | - Jeremy M Adelstein
- From the College of Nursing (J.M.T., J.M.A., L.E.W.), The Ohio State University, Columbus
| | | | - Colin K Combs
- Department of Biomedical Sciences, University of North Dakota, Grand Forks (C.K.C.)
| | - Loren E Wold
- From the College of Nursing (J.M.T., J.M.A., L.E.W.), The Ohio State University, Columbus
- Department of Physiology and Cell Biology, College of Medicine (L.E.W.), The Ohio State University, Columbus
| |
Collapse
|
36
|
Umar T, Shalini S, Raza MK, Gusain S, Kumar J, Ahmed W, Tiwari M, Hoda N. New amyloid beta-disaggregating agents: synthesis, pharmacological evaluation, crystal structure and molecular docking of N-(4-((7-chloroquinolin-4-yl)oxy)-3-ethoxybenzyl)amines. MEDCHEMCOMM 2018; 9:1891-1904. [PMID: 30568757 PMCID: PMC6254049 DOI: 10.1039/c8md00312b] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 08/06/2018] [Indexed: 11/21/2022]
Abstract
In the journey towards the development of potent multi-targeted ligands for the treatment of Alzheimer's disease, a series of Aβ aggregation inhibitors having quinoline scaffold were designed utilizing computational biology tools, synthesized and characterized by various spectral techniques including single-crystal X-ray crystallography. Organic syntheses relying upon convergent synthetic routes were employed. Investigations via ThT fluorescence assay, electron microscopy and transmission electron microscopy revealed the synthesized derivatives to exhibit Aβ self-aggregation inhibition. Molecules 5g and 5a showed the highest inhibitory potential, 53.73% and 53.63% at 50 μM respectively; higher than the standard Aβ disaggregating agent, curcumin. Molecules 5g and 5a disaggregated AChE-induced (58.26%, 47.36%) Aβ aggregation more than two fold more than the standard drug-donepezil (23.66%) and inhibited Cu2+-induced Aβ aggregation. A docking study significantly showed their interaction with key residues of Aβ and the results were in accordance with the study. Besides, these compounds also exhibited potential antioxidant activity (5a, 2.7240 Trolox equivalent by ORAC assay) and metal chelating property. Furthermore, the stoichiometric ratio of Cu (ii)-5a and Cu(ii)-5g complexes were found by Job's method (0.5 : 1 for 5a and 0.8 : 1 for 5g). In silico ADMET profiling showed these derivatives to have drug like properties with very low toxicity effects in the pharmacokinetic study. Overall, these results displayed a multi-activity profile with promising Aβ aggregation inhibition and antioxidation and metal chelation activity that could be helpful for developing new multifunctional agents against Alzheimer's disease.
Collapse
Affiliation(s)
- Tarana Umar
- Department of Chemistry , Jamia Millia Islamia (Central University) , New Delhi 110025 , India .
| | - Shruti Shalini
- Dr. B. R. Ambedkar Centre for Biomedical Research , University of Delhi , New Delhi 110007 , India .
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry , Indian Institute of Science , Bangalore 560012 , India
| | - Siddharth Gusain
- Dr. B. R. Ambedkar Centre for Biomedical Research , University of Delhi , New Delhi 110007 , India .
| | - Jitendra Kumar
- Department of Chemistry , Sardar Vallabhbhai Patel College , Kaimur- 821101, V. K. S. U., Ara , Bhabua , Bihar-802301 , India
| | - Waqar Ahmed
- Department of Chemistry , Jamia Millia Islamia (Central University) , New Delhi 110025 , India .
| | - Manisha Tiwari
- Dr. B. R. Ambedkar Centre for Biomedical Research , University of Delhi , New Delhi 110007 , India .
| | - Nasimul Hoda
- Department of Chemistry , Jamia Millia Islamia (Central University) , New Delhi 110025 , India .
| |
Collapse
|
37
|
Chen H, Su F, Ye Q, Wang Z, Shu H, Bai F. The Dose-Dependent Effects of Vascular Risk Factors on Dynamic Compensatory Neural Processes in Mild Cognitive Impairment. Front Aging Neurosci 2018; 10:131. [PMID: 29867442 PMCID: PMC5951955 DOI: 10.3389/fnagi.2018.00131] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/20/2018] [Indexed: 01/14/2023] Open
Abstract
Background/Objectives: Mild cognitive impairment (MCI) has been associated with risk for Alzheimer's Disease (AD). Previous investigations have suggested that vascular risk factors (VRFs) were associated with cognitive decline and AD pathogenesis, and the intervention of VRFs may be a possible way to prevent dementia. However, in MCI, little is known about the potential impacts of VRFs on neural networks and their neural substrates, which may be a neuroimaging biomarker of the disease progression. Methods: 128 elderly Han Chinese participants (67 MCI subjects and 61 matched normal elderly) with or without VRFs (hypertension, diabetes mellitus, hypercholesterolemia, smoking and alcohol drinking) underwent the resting-state functional magnetic resonance imaging (fMRI) and neuropsychological tests. We obtained the default mode network (DMN) to identify alterations in MCI with the varying number of the VRF and analyzed the significant correlation with behavioral performance. Results: The effects of VRF on the DMN were primarily in bilateral dorsolateral prefrontal cortex (DLPFC) (i.e., middle frontal gyrus). Normal elderly showed the gradually increased functional activity of DLPFC, while a fluctuant activation of DLPFC was displayed in MCI with the growing number of the VRF. Interestingly, the left DLPFC further displayed significantly dynamic correlation with executive function as the variation of VRF loading. Initial level of compensation was observed in normal aging and none-vascular risk factor (NVRF) MCI, while these compensatory neural processes were suppressed in One-VRF MCI and were subsequently re-aroused in Over-One-VRF MCI. Conclusions: These findings suggested that the dose-dependent effects of VRF on DLPFC were highlighted in MCI, and the dynamic compensatory neural processes that fluctuated along with variations of VRF loading could be key role in the progression of MCI.
Collapse
Affiliation(s)
- Haifeng Chen
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Fan Su
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Qing Ye
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zan Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hao Shu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Feng Bai
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
38
|
Zhan X, Stamova B, Sharp FR. Lipopolysaccharide Associates with Amyloid Plaques, Neurons and Oligodendrocytes in Alzheimer's Disease Brain: A Review. Front Aging Neurosci 2018. [PMID: 29520228 PMCID: PMC5827158 DOI: 10.3389/fnagi.2018.00042] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This review proposes that lipopolysaccharide (LPS, found in the wall of all Gram-negative bacteria) could play a role in causing sporadic Alzheimer’s disease (AD). This is based in part upon recent studies showing that: Gram-negative E. coli bacteria can form extracellular amyloid; bacterial-encoded 16S rRNA is present in all human brains with over 70% being Gram-negative bacteria; ultrastructural analyses have shown microbes in erythrocytes of AD patients; blood LPS levels in AD patients are 3-fold the levels in control; LPS combined with focal cerebral ischemia and hypoxia produced amyloid-like plaques and myelin injury in adult rat cortex. Moreover, Gram-negative bacterial LPS was found in aging control and AD brains, though LPS levels were much higher in AD brains. In addition, LPS co-localized with amyloid plaques, peri-vascular amyloid, neurons, and oligodendrocytes in AD brains. Based upon the postulate LPS caused oligodendrocyte injury, degraded Myelin Basic Protein (dMBP) levels were found to be much higher in AD compared to control brains. Immunofluorescence showed that the dMBP co-localized with β amyloid (Aβ) and LPS in amyloid plaques in AD brain, and dMBP and other myelin molecules were found in the walls of vesicles in periventricular White Matter (WM). These data led to the hypothesis that LPS acts on leukocyte and microglial TLR4-CD14/TLR2 receptors to produce NFkB mediated increases of cytokines which increase Aβ levels, damage oligodendrocytes and produce myelin injury found in AD brain. Since Aβ1–42 is also an agonist for TLR4 receptors, this could produce a vicious cycle that accounts for the relentless progression of AD. Thus, LPS, the TLR4 receptor complex, and Gram-negative bacteria might be treatment or prevention targets for sporadic AD.
Collapse
Affiliation(s)
- Xinhua Zhan
- Department of Neurology, MIND Institute, University of California, Davis, Davis, CA, United States
| | - Boryana Stamova
- Department of Neurology, MIND Institute, University of California, Davis, Davis, CA, United States
| | - Frank R Sharp
- Department of Neurology, MIND Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
39
|
Kosenko EA, Tikhonova LA, Montoliu C, Barreto GE, Aliev G, Kaminsky YG. Metabolic Abnormalities of Erythrocytes as a Risk Factor for Alzheimer's Disease. Front Neurosci 2018; 11:728. [PMID: 29354027 PMCID: PMC5760569 DOI: 10.3389/fnins.2017.00728] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/13/2017] [Indexed: 01/02/2023] Open
Abstract
Alzheimer's disease (AD) is a slowly progressive, neurodegenerative disorder of uncertain etiology. According to the amyloid cascade hypothesis, accumulation of non-soluble amyloid β peptides (Aβ) in the Central Nervous System (CNS) is the primary cause initiating a pathogenic cascade leading to the complex multilayered pathology and clinical manifestation of the disease. It is, therefore, not surprising that the search for mechanisms underlying cognitive changes observed in AD has focused exclusively on the brain and Aβ-inducing synaptic and dendritic loss, oxidative stress, and neuronal death. However, since Aβ depositions were found in normal non-demented elderly people and in many other pathological conditions, the amyloid cascade hypothesis was modified to claim that intraneuronal accumulation of soluble Aβ oligomers, rather than monomer or insoluble amyloid fibrils, is the first step of a fatal cascade in AD. Since a characteristic reduction of cerebral perfusion and energy metabolism occurs in patients with AD it is suggested that capillary distortions commonly found in AD brain elicit hemodynamic changes that alter the delivery and transport of essential nutrients, particularly glucose and oxygen to neuronal and glial cells. Another important factor in tissue oxygenation is the ability of erythrocytes (red blood cells, RBC) to transport and deliver oxygen to tissues, which are first of all dependent on the RBC antioxidant and energy metabolism, which finally regulates the oxygen affinity of hemoglobin. In the present review, we consider the possibility that metabolic and antioxidant defense alterations in the circulating erythrocyte population can influence oxygen delivery to the brain, and that these changes might be a primary mechanism triggering the glucose metabolism disturbance resulting in neurobiological changes observed in the AD brain, possibly related to impaired cognitive function. We also discuss the possibility of using erythrocyte biochemical aberrations as potential tools that will help identify a risk factor for AD.
Collapse
Affiliation(s)
- Elena A Kosenko
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Lyudmila A Tikhonova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Carmina Montoliu
- Fundación Investigación Hospital Clínico, INCLIVA Instituto Investigación Sanitaria, Valencia, Spain
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Gjumrakch Aliev
- GALLY International Biomedical Research Institute Inc., San Antonio, TX, United States
| | - Yury G Kaminsky
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|
40
|
Hemanth Kumar B, Dinesh Kumar B, Diwan PV. Hesperidin, a citrus flavonoid, protects against l-methionine-induced hyperhomocysteinemia by abrogation of oxidative stress, endothelial dysfunction and neurotoxicity in Wistar rats. PHARMACEUTICAL BIOLOGY 2017; 55:146-155. [PMID: 27677544 PMCID: PMC7011910 DOI: 10.1080/13880209.2016.1231695] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 08/30/2016] [Indexed: 06/06/2023]
Abstract
CONTEXT Hesperidin (HSP), a flavanoglycone found in citrus fruits, has antioxidant, anti-inflammatory and neuroprotective properties. OBJECTIVE This study evaluates the protective effect of HSP on l-methionine-induced hyperhomocysteinemia (HHcy) in rats. MATERIALS AND METHODS Male Wistar rats were randomly divided into seven groups as DMSO, l-methionine, HSP (25, 50 and 100 mg/kg), HSP-per se (100 mg/kg) and donepezil (0.1 mg/kg). HHcy was induced by oral administration of l-methionine (1.7 g/kg) for 32 days. From the 14th day of study HSP (25, 50 and 100 mg/kg) and donepezil was administered orally to l-methionine-treated rats. Cognitive impairment induced by HHcy was determined using the Morris water maze (MWM) and Y-maze on video tracking system (28th-32nd day). Different biomarkers of HHcy in serum and brain and vascular reactivity were evaluated and histopathology (thoracic aorta and brain) was done. RESULTS HSP (100 mg/kg) treatment in l-methionine-treated rats exhibited significant (p < 0.001) dose-dependent activity and reduced behavioural deficits, brain acetylcholinesterase (25.99 ± 2.36 versus 10.73 ± 1.26 μmoles/mg), brain lipid peroxidation (15.25 ± 1.65 versus 6.18 ± 0.74 nM/mg), serum homocysteine (Hcy) (22.37 ± 0.30 versus 11.01 ± 1.01 μg/mL) and serum cholesterol (182.7 ± 2.15 versus 101.5 ± 2.76 mg/dL) and increased brain antioxidant levels. HSP significantly (p < 0.001) reduced endothelial dysfunction (ED) by abolishing the effect of l-methionine on acetylcholine-induced endothelial-dependent relaxation and increased serum nitrite and vascular nitric oxide bioavailability along with the restoration of histological aberrations. CONCLUSION HSP exerts a protective effect on HHcy by abrogating oxidative stress, ED and neurotoxicity.
Collapse
Affiliation(s)
- B. Hemanth Kumar
- Department of Pharmacology, Pharmacology, Anurag Group of Institutions (Formerly Lalitha College of Pharmacy), Hyderabad, Telangana, India
- Pharmaceutical Sciences, Research and Development cell, Jawaharlal Nehru Technological University, Kukatpally, Hyderabad, Telangana, India
| | - B. Dinesh Kumar
- Food and Drug Toxicology Research Centre (FDTRC), National Institute of Nutrition (ICMR), Hyderabad, Telangana, India
| | - Prakash V. Diwan
- Department of Pharmacology, Pharmacology, Anurag Group of Institutions (Formerly Lalitha College of Pharmacy), Hyderabad, Telangana, India
- Maratha Mandal Research Center, Belgaum, Karnataka, India
| |
Collapse
|
41
|
Yuan XY, Wang XG. Mild cognitive impairment in type 2 diabetes mellitus and related risk factors: a review. Rev Neurosci 2017; 28:715-723. [DOI: 10.1515/revneuro-2017-0016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/18/2017] [Indexed: 01/05/2023]
Abstract
AbstractType 2 diabetes mellitus (T2DM) is a global epidemic disease and has become a significant health problem. Many studies have raised concern about the mild cognitive impairment (MCI) with T2DM and even the Alzheimer’s disease patients with T2DM. The incidence of MCI is higher in individuals with T2DM than those without diabetes. Cognitive changes might affect everyday activities depending on the work and situation. Although the exact pathophysiology of MCI in T2DM is unclear, many studies suggest that the alterations in pathoglycemia, diabetic complications, related end products, and physical/psychological status are significant risk factors. In this article, we systematically overview the studies to illustrate the related risk factors of cognitive impairment in patients with T2DM. Further high quality studies and treatment need to be initiated and it will become incumbent on clinicians to identify and cure the earliest signs of clinical impairment.
Collapse
Affiliation(s)
- Xiao-Ying Yuan
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian 116044, P.R. China
| | - Xu-Gang Wang
- Department of Neurology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, P.R. China
| |
Collapse
|
42
|
Curcumin Reverses the Diazepam-Induced Cognitive Impairment by Modulation of Oxidative Stress and ERK 1/2/NF- κB Pathway in Brain. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3037876. [PMID: 29098059 PMCID: PMC5643119 DOI: 10.1155/2017/3037876] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022]
Abstract
Oxidative stress and inflammation can be involved in cognitive dysfunction associated with neurodegenerative disorders. Diazepam (DZP) administration has been chosen to simulate the memory impairment. The aim of this study was to evaluate the effects of curcumin (CUR) on spatial cognition, ambulatory activity, and blood and brain oxidative stress levels. The ERK/NF-κB signaling pathway and the histopathological changes in the hippocampus and frontal lobe, in diazepam-treated rats, were also analyzed. The animals were divided into 4 groups: control, carboxymethylcellulose (CMC) + CUR, CMC + DZP, and CUR + CMC + DZP. CUR (150 mg/kg b.w.) was orally administered for 28 days. DZP (2 mg/kg b.w.) was intraperitoneally administered 20 minutes before the behavioral tests (open field test, Y-maze, and elevated plus maze). CUR improved the spontaneous alternation behavior, decreased the oxidative stress levels, both in the blood and in the hippocampus, and downregulated the extracellular signal-regulated kinase (ERK 1/2)/nuclear transcription factor- (NF-) κB/pNF-κB pathway in the hippocampus and the iNOS expression in the hippocampus and frontal lobe of the DZP-treated rats. Histopathologically, no microscopic changes were found. The immunohistochemical signal of iNOS decreased in the DZP and CUR-treated group. Thus, our findings suggest that curcumin administration may improve the cognitive performance and may also have an antioxidant effect.
Collapse
|
43
|
Rojas-Gutierrez E, Muñoz-Arenas G, Treviño S, Espinosa B, Chavez R, Rojas K, Flores G, Díaz A, Guevara J. Alzheimer's disease and metabolic syndrome: A link from oxidative stress and inflammation to neurodegeneration. Synapse 2017. [PMID: 28650104 DOI: 10.1002/syn.21990] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and one of the most important causes of morbidity and mortality among the aging population. AD diagnosis is made post-mortem, and the two pathologic hallmarks, particularly evident in the end stages of the illness, are amyloid plaques and neurofibrillary tangles. Currently, there is no curative treatment for AD. Additionally, there is a strong relation between oxidative stress, metabolic syndrome, and AD. The high levels of circulating lipids and glucose imbalances amplify lipid peroxidation that gradually diminishes the antioxidant systems, causing high levels of oxidative metabolism that affects cell structure, leading to neuronal damage. Accumulating evidence suggests that AD is closely related to a dysfunction of both insulin signaling and glucose metabolism in the brain, leading to an insulin-resistant brain state. Four drugs are currently used for this pathology: Three FDA-approved cholinesterase inhibitors and one NMDA receptor antagonist. However, wide varieties of antioxidants are promissory to delay or prevent the symptoms of AD and may help in treating the disease. Therefore, therapeutic efforts to achieve attenuation of oxidative stress could be beneficial in AD treatment, attenuating Aβ-induced neurotoxicity and improve neurological outcomes in AD. The term inflammaging characterizes a widely accepted paradigm that aging is accompanied by a low-grade chronic up-regulation of certain pro-inflammatory responses in the absence of overt infection, and is a highly significant risk factor for both morbidity and mortality in the elderly.
Collapse
Affiliation(s)
- Eduardo Rojas-Gutierrez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Guadalupe Muñoz-Arenas
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Samuel Treviño
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Blanca Espinosa
- Departamento de Bioquímica, Instituto Nacional de Enfermedades Respiratorias-INER, Ciudad de México, Mexico
| | - Raúl Chavez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Karla Rojas
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Alfonso Díaz
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Jorge Guevara
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
44
|
Borai IH, Ezz MK, Rizk MZ, Aly HF, El-Sherbiny M, Matloub AA, Fouad GI. Therapeutic impact of grape leaves polyphenols on certain biochemical and neurological markers in AlCl 3-induced Alzheimer's disease. Biomed Pharmacother 2017; 93:837-851. [PMID: 28715867 DOI: 10.1016/j.biopha.2017.07.038] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 11/28/2022] Open
Abstract
Alzheimer's disease (AD) is a grave and prevailing neurodegenerative disease, characterized by slow and progressive neurodegeneration in different brain regions. Aluminum (Al) is a potent and widely distributed neurotoxic metal, implicated in the neuropathogenesis of AD. This study aimed to evaluate the possible neurorestorative potential of Vitis vinifera Leaves Polyphenolic (VLP) extract in alleviating aluminum chloride (AlCl3)-induced neurotoxicity in male rats. AlCl3 neurotoxicity induced a significant decrease in brain/serum acetylcholine (ACh) contents and serum dopamine (DA) levels, along with a significant increment of brain/serum acetylcholinesterase (AChE) activities. In addition, Al treatment resulted in significantly decreased serum levels of both total antioxidant capacity (TAC) and brain-derived neurotrophic factor (BDNF), and significantly increased serum levels of both interleukin-6 (IL-6) and total homocysteine (tHcy), as compared to control. Behavioral alterations, assessed by the T-maze test, showed impaired cognitive function. Furthermore, AD-brains revealed an increase in DNA fragmentation as evidenced by comet assay. AlCl3 induction also caused histopathological alterations in AD-brain. Treatment of AD-rats with VLP extract (100mg/kg body weight/day) improved neurobehavioral changes, as evidenced by the improvement in brain function, as well as, modulation of most biochemical markers, and confirmed by T-maze test, the histopathological study of the brain and comet assay. The current work indicates that the VLP extract has neuroprotective, antioxidative, anti-inflammatory, and anti-amnesic activities against AlCl3-induced cerebral damages and neurocognitive dysfunction.
Collapse
Affiliation(s)
- Ibrahim H Borai
- Biochemistry Department, Faculty of Science, Ain-Shams University, Cairo, Egypt.
| | - Magda K Ezz
- Biochemistry Department, Faculty of Science, Ain-Shams University, Cairo, Egypt.
| | - Maha Z Rizk
- Therapeutical Chemistry Department, National Research Center, Dokki, Cairo, Egypt.
| | - Hanan F Aly
- Therapeutical Chemistry Department, National Research Center, Dokki, Cairo, Egypt.
| | - Mahmoud El-Sherbiny
- Therapeutical Chemistry Department, National Research Center, Dokki, Cairo, Egypt.
| | - Azza A Matloub
- Pharmacognosy Department, National Research Center, Dokki, Cairo, Egypt.
| | - Ghadha I Fouad
- Therapeutical Chemistry Department, National Research Center, Dokki, Cairo, Egypt.
| |
Collapse
|
45
|
Freitas HR, Isaac AR, Malcher-Lopes R, Diaz BL, Trevenzoli IH, De Melo Reis RA. Polyunsaturated fatty acids and endocannabinoids in health and disease. Nutr Neurosci 2017; 21:695-714. [PMID: 28686542 DOI: 10.1080/1028415x.2017.1347373] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) are lipid derivatives of omega-3 (docosahexaenoic acid, DHA, and eicosapentaenoic acid, EPA) or of omega-6 (arachidonic acid, ARA) synthesized from membrane phospholipids and used as a precursor for endocannabinoids (ECs). They mediate significant effects in the fine-tune adjustment of body homeostasis. Phyto- and synthetic cannabinoids also rule the daily life of billions worldwide, as they are involved in obesity, depression and drug addiction. Consequently, there is growing interest to reveal novel active compounds in this field. Cloning of cannabinoid receptors in the 90s and the identification of the endogenous mediators arachidonylethanolamide (anandamide, AEA) and 2-arachidonyglycerol (2-AG), led to the characterization of the endocannabinoid system (ECS), together with their metabolizing enzymes and membrane transporters. Today, the ECS is known to be involved in diverse functions such as appetite control, food intake, energy balance, neuroprotection, neurodegenerative diseases, stroke, mood disorders, emesis, modulation of pain, inflammatory responses, as well as in cancer therapy. Western diet as well as restriction of micronutrients and fatty acids, such as DHA, could be related to altered production of pro-inflammatory mediators (e.g. eicosanoids) and ECs, contributing to the progression of cardiovascular diseases, diabetes, obesity, depression or impairing conditions, such as Alzheimer' s disease. Here we review how diets based in PUFAs might be linked to ECS and to the maintenance of central and peripheral metabolism, brain plasticity, memory and learning, blood flow, and genesis of neural cells.
Collapse
Affiliation(s)
- Hércules Rezende Freitas
- a Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| | - Alinny Rosendo Isaac
- a Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| | | | - Bruno Lourenço Diaz
- c Laboratory of Inflammation, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| | - Isis Hara Trevenzoli
- d Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| | - Ricardo Augusto De Melo Reis
- a Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho , Universidade Federal do Rio de Janeiro, Cidade Universitária , Ilha do Fundão, Rio de Janeiro , RJ 21941-902 , Brazil
| |
Collapse
|
46
|
Takane K, Hasegawa Y, Lin B, Koibuchi N, Cao C, Yokoo T, Kim-Mitsuyama S. Detrimental Effects of Centrally Administered Angiotensin II are Enhanced in a Mouse Model of Alzheimer Disease Independently of Blood Pressure. J Am Heart Assoc 2017; 6:JAHA.116.004897. [PMID: 28428194 PMCID: PMC5533006 DOI: 10.1161/jaha.116.004897] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background The significance of brain angiotensin II in Alzheimer disease (AD) is unclear. Methods and Results To examine the role of brain angiotensin II in AD, intracerebroventricular angiotensin II infusion was performed on 5XFAD mice, a mouse model of AD, and wild‐type mice, and the detrimental effects of brain angiotensin II was compared between the 2 strains of mice. Intracerebroventricular angiotensin II infusion significantly impaired cognitive function in 5XFAD mice but not in wild‐type mice. This vulnerability of 5XFAD mice to brain angiotensin II was associated with enhancement of hippocampal inflammation and oxidative stress and with increased cerebrovascular amyloid β deposition. We also compared the effect of brain angiotensin II on the heart and skeletal muscle between the 2 strains because AD is associated with heart failure and sarcopenia. We found that cardiac compensatory response of 5XFAD mice to brain angiotensin II–induced hypertension was less than that of wild‐type mice. Brain angiotensin II caused skeletal muscle atrophy and injury in 5XFAD mice more than in wild‐type mice. Conclusions Brain angiotensin II seems to be involved in cognitive impairment and brain injury in AD, which is associated with oxidative stress, inflammation, and cerebral amyloid angiopathy. Further, brain angiotensin II may participate in cardiac disease and sarcopenia observed in AD.
Collapse
Affiliation(s)
- Koki Takane
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan.,Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Yu Hasegawa
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Bowen Lin
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Nobutaka Koibuchi
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Cheng Cao
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Shokei Kim-Mitsuyama
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| |
Collapse
|
47
|
Badhwar A, Brown R, Stanimirovic DB, Haqqani AS, Hamel E. Proteomic differences in brain vessels of Alzheimer's disease mice: Normalization by PPARγ agonist pioglitazone. J Cereb Blood Flow Metab 2017; 37:1120-1136. [PMID: 27339263 PMCID: PMC5363486 DOI: 10.1177/0271678x16655172] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cerebrovascular insufficiency appears years prior to clinical symptoms in Alzheimer's disease. The soluble, highly toxic amyloid-β species, generated from the amyloidogenic processing of amyloid precursor protein, are known instigators of the chronic cerebrovascular insufficiency observed in both Alzheimer's disease patients and transgenic mouse models. We have previously demonstrated that pioglitazone potently reverses cerebrovascular impairments in a mouse model of Alzheimer's disease overexpressing amyloid-β. In this study, we sought to characterize the effects of amyloid-β overproduction on the cerebrovascular proteome; determine how pioglitazone treatment affected the altered proteome; and analyze the relationship between normalized protein levels and recovery of cerebrovascular function. Three-month-old wildtype and amyloid precursor protein mice were treated with pioglitazone- (20 mg/kg/day, 14 weeks) or control-diet. Cerebral arteries were surgically isolated, and extracted proteins analyzed by gel-free and gel-based mass spectrometry. 193 cerebrovascular proteins were abnormally expressed in amyloid precursor protein mice. Pioglitazone treatment rescued a third of these proteins, mainly those associated with oxidative stress, promotion of cerebrovascular vasocontractile tone, and vascular compliance. Our results demonstrate that amyloid-β overproduction perturbs the cerebrovascular proteome. Recovery of cerebrovascular function with pioglitazone is associated with normalized levels of key proteins in brain vessel function, suggesting that pioglitazone-responsive cerebrovascular proteins could be early biomarkers of Alzheimer's disease.
Collapse
Affiliation(s)
- AmanPreet Badhwar
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Rebecca Brown
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Danica B Stanimirovic
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Arsalan S Haqqani
- Human Health Therapeutics, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
- Edith Hamel, Laboratory of Cerebrovascular research, Montreal Neurological Institute, 3801 University St., Montréal, QC, H3A 2B4, Canada.
| |
Collapse
|
48
|
Mamelak M. Energy and the Alzheimer brain. Neurosci Biobehav Rev 2017; 75:297-313. [PMID: 28193453 DOI: 10.1016/j.neubiorev.2017.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 01/01/2023]
Abstract
The high energy demands of the poorly myelinated long axon hippocampal and cortical neurons render these neurons selectively vulnerable to degeneration in Alzheimer's disease. However, pathology engages all of the major elements of the neurovascular unit of the mature Alzheimer brain, the neurons, glia and blood vessels. Neurons present with retrograde degeneration of the axodendritic tree, capillaries with string vessels and markedly reduced densities and glia with signs of inflammatory activation. The neurons, capillaries and astrocytes of the mature Alzheimer brain harbor structurally defective mitochondria. Clinically, reduced glucose utilization, decades before cognitive deterioration, betrays ongoing energy insufficiency. β-hydroxybutyrate and γ-hydroxybutyrate can both provide energy to the brain when glucose utilization is blocked. Early work in mouse models of Alzheimer's disease demonstrate their ability to reverse the pathological changes in the Alzheimer brain and initial clinical trials reveal their ability to improve cognition and every day function. Supplying the brain with energy holds great promise for delaying the onset of Alzheimer's disease and slowing its progress.
Collapse
|
49
|
Nakagawa T, Hasegawa Y, Uekawa K, Senju S, Nakagata N, Matsui K, Kim-Mitsuyama S. Transient Mild Cerebral Ischemia Significantly Deteriorated Cognitive Impairment in a Mouse Model of Alzheimer's Disease via Angiotensin AT1 Receptor. Am J Hypertens 2017; 30:141-150. [PMID: 27572961 DOI: 10.1093/ajh/hpw099] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 07/21/2016] [Accepted: 08/03/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Ischemic stroke is suggested to be potentially associated with cognitive impairment in Alzheimer's disease (AD). We hypothesized that cerebral ischemia deteriorates cognitive impairment in AD, through angiotensin II. METHODS We used 5XFAD mouse, a model of AD with vascular and cerebral amyloid-β deposition. Transient cerebral ischemia of mice was induced by bilateral common carotid artery occlusion (BCCAO) for 17 minutes. The posttreatment with olmesartan, an ARB, or vehicle was started at 24 hours after BCCAO and was performed for 5 weeks. Experimental mice consisted of 5 groups: (i) wild-type mice, (ii) wild-type mice with BCCAO, (iii) 5XFAD mice, (iv) 5XFAD mice with BCCAO, (v) 5XFAD mice with BCCAO and olmesartan postadministration. RESULTS BCCAO in 5XFAD caused greater escape latency (P < 0.01) on water maze test than that in wild type, indicating that transient brief cerebral ischemia enhanced cognitive decline in 5XFAD mice. Posttreatment with olmesartan significantly reduced escape latency (P < 0.01) on water maze test, retention trial latency (P < 0.05) on passive avoidance test, and retention time of outer zone (P < 0.01) on open-field test in 5XFAD subjected to BCCAO. This protective effect of olmesartan against cognitive impairment in 5XFAD with BCCAO was associated with the protection of neuron and attenuation of oxidative stress in hippocampus and the suppression of blood-brain barrier disruption. CONCLUSIONS We obtained the evidence that transient brief cerebral ischemia deteriorated cognitive impairment in AD model through AT1 receptor.
Collapse
Affiliation(s)
- Takashi Nakagawa
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Yu Hasegawa
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Ken Uekawa
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Kumamoto, Japan
| | - Kunihiko Matsui
- Department of General and Community Medicine, Kumamoto University Hospital, Kumamoto, Japan
| | - Shokei Kim-Mitsuyama
- Departments of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan;
| |
Collapse
|
50
|
Khan MS, Misra SK, Wang Z, Daza E, Schwartz-Duval AS, Kus JM, Pan D, Pan D. Paper-Based Analytical Biosensor Chip Designed from Graphene-Nanoplatelet-Amphiphilic-diblock-co-Polymer Composite for Cortisol Detection in Human Saliva. Anal Chem 2017; 89:2107-2115. [DOI: 10.1021/acs.analchem.6b04769] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Muhammad S. Khan
- Biomedical
Research Center, Carle Foundation Hospital, Urbana, Illinois United States
| | - Santosh K. Misra
- Biomedical
Research Center, Carle Foundation Hospital, Urbana, Illinois United States
| | - Zhen Wang
- Biomedical
Research Center, Carle Foundation Hospital, Urbana, Illinois United States
| | - Enrique Daza
- Biomedical
Research Center, Carle Foundation Hospital, Urbana, Illinois United States
| | | | - Joseph M. Kus
- Biomedical
Research Center, Carle Foundation Hospital, Urbana, Illinois United States
| | | | - Dipanjan Pan
- Biomedical
Research Center, Carle Foundation Hospital, Urbana, Illinois United States
| |
Collapse
|