1
|
Alami M, Boumezough K, Zerif E, Zoubdane N, Khalil A, Bunt T, Laurent B, Witkowski JM, Ramassamy C, Boulbaroud S, Fulop T, Berrougui H. In Vitro Assessment of the Neuroprotective Effects of Pomegranate ( Punica granatum L.) Polyphenols Against Tau Phosphorylation, Neuroinflammation, and Oxidative Stress. Nutrients 2024; 16:3667. [PMID: 39519499 PMCID: PMC11547808 DOI: 10.3390/nu16213667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Oxidative stress and chronic inflammation, at both the systemic and the central level, are critical early events in atherosclerosis and Alzheimer's disease (AD). PURPOSE To investigate the oxidative stress-, inflammation-, and Tau-phosphorylation-lowering effects of pomegranate polyphenols (PPs) (punicalagin, ellagic acid, peel, and aril extracts). METHODS We used flow cytometry to quantify the protein expression of proinflammatory cytokines (IL-1β) and anti-inflammatory mediators (IL-10) in THP-1 macrophages, as well as M1/M2 cell-specific marker (CD86 and CD163) expression in human microglia HMC3 cells. The IL-10 protein expression was also quantified in U373-MG human astrocytes. The effect of PPs on human amyloid beta 1-42 (Aβ1-42)-induced oxidative stress was assessed in the microglia by measuring ROS generation and lipid peroxidation, using 2',7'-dichlorofluorescein diacetate (DCFH-DA) and thiobarbituric acid reactive substance (TBARS) tests, respectively. Neuronal viability and cell apoptotic response to Aβ1-42 toxicity were assayed using the MTT (3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide) assay and the annexin-V-FITC apoptosis detection kit, respectively. Finally, flow cytometry analysis was also performed to evaluate the ability of PPs to modulate Aβ1-42-induced Tau-181 phosphorylation (pTau-181). RESULTS Our data indicate that PPs are significantly (p < 0.05) effective in countering Aβ1-42-induced inflammation through increasing the anti-inflammatory cytokines (IL-10) in U373-MG astrocytes and THP1 macrophages and decreasing proinflammatory marker (IL-1β) expression in THP1 macrophages. The PPs were also significantly (p < 0.05) effective in inducing the phenotypic transition of THP-1 macrophages and microglial cells from M1 to M2 by decreasing CD86 and increasing CD163 surface receptor expression. Moreover, our treatments have a significant (p < 0.05) beneficial impact on oxidative stress, illustrated in the reduction in TBARS and ROS generation. Our treatments have significant (p < 0.05) cell viability improvement capacities and anti-apoptotic effects on human H4 neurons. Furthermore, our results suggest that Aβ1-42 significantly (p < 0.05) increases pTau-181. This effect is significantly (p < 0.05) attenuated by arils, peels, and punicalagin and drastically reduced by the ellagic acid treatment. CONCLUSION Overall, our results attribute to PPs anti-inflammatory, antioxidant, anti-apoptotic, and anti-Tau-pathology potential. Future studies should aim to extend our knowledge of the potential role of PPs in Aβ1-42-induced neurodegeneration, particularly concerning its association with the tauopathy involved in AD.
Collapse
Affiliation(s)
- Mehdi Alami
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal 23020, Morocco; (M.A.); (K.B.); (S.B.)
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada; (E.Z.); (N.Z.); (A.K.); (B.L.); (T.F.)
| | - Kaoutar Boumezough
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal 23020, Morocco; (M.A.); (K.B.); (S.B.)
| | - Echarki Zerif
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada; (E.Z.); (N.Z.); (A.K.); (B.L.); (T.F.)
| | - Nada Zoubdane
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada; (E.Z.); (N.Z.); (A.K.); (B.L.); (T.F.)
| | - Abdelouahed Khalil
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada; (E.Z.); (N.Z.); (A.K.); (B.L.); (T.F.)
| | - Ton Bunt
- Izumi Biosciences, Inc., Lexington, MA 02420, USA;
| | - Benoit Laurent
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada; (E.Z.); (N.Z.); (A.K.); (B.L.); (T.F.)
| | - Jacek M. Witkowski
- Department of Embryology, Medical University of Gdansk, 80-210 Gdańsk, Poland;
| | - Charles Ramassamy
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC H7V 1B7, Canada;
| | - Samira Boulbaroud
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal 23020, Morocco; (M.A.); (K.B.); (S.B.)
| | - Tamas Fulop
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada; (E.Z.); (N.Z.); (A.K.); (B.L.); (T.F.)
| | - Hicham Berrougui
- Department of Biology, Polydisciplinary Faculty, University Sultan Moulay Slimane, Beni Mellal 23020, Morocco; (M.A.); (K.B.); (S.B.)
- Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 4N4, Canada; (E.Z.); (N.Z.); (A.K.); (B.L.); (T.F.)
| |
Collapse
|
2
|
Pluta R, Miziak B, Czuczwar SJ. Post-Ischemic Permeability of the Blood-Brain Barrier to Amyloid and Platelets as a Factor in the Maturation of Alzheimer's Disease-Type Brain Neurodegeneration. Int J Mol Sci 2023; 24:10739. [PMID: 37445917 DOI: 10.3390/ijms241310739] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/13/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
The aim of this review is to present evidence of the impact of ischemic changes in the blood-brain barrier on the maturation of post-ischemic brain neurodegeneration with features of Alzheimer's disease. Understanding the processes involved in the permeability of the post-ischemic blood-brain barrier during recirculation will provide clinically relevant knowledge regarding the neuropathological changes that ultimately lead to dementia of the Alzheimer's disease type. In this review, we try to distinguish between primary and secondary neuropathological processes during and after ischemia. Therefore, we can observe two hit stages that contribute to Alzheimer's disease development. The onset of ischemic brain pathology includes primary ischemic neuronal damage and death followed by the ischemic injury of the blood-brain barrier with serum leakage of amyloid into the brain tissue, leading to increased ischemic neuronal susceptibility to amyloid neurotoxicity, culminating in the formation of amyloid plaques and ending in full-blown dementia of the Alzheimer's disease type.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
3
|
Ye X, Chen J, Pan J, Wu Q, Wang Y, Lu M, Zhang C, Zhang Z, Ma M, Zhu J, Vella AT, Wan J, Wang K. Interleukin-17 Promotes the Infiltration of CD8+ T Cells into the Brain in a Mouse Model for Alzheimer's Disease. Immunol Invest 2023; 52:135-153. [PMID: 36394561 DOI: 10.1080/08820139.2022.2136525] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Interleukin-17 (IL-17) family cytokines play critical roles in inflammation and pathogen resistance. Inflammation in the central nervous system, denoted as neuroinflammation, promotes the onset and progression of Alzheimer's disease (AD). Previous studies showed that IL-17A neutralizing antibody treatment alleviated Amyloid β (Aβ) burden in rodent models of AD, while overexpression of IL-17A in mouse lateral ventricles rescued part of the AD pathology. However, the involvement of IL-17 in AD and its mechanism of action remain largely unknown. METHODS To investigate the role of IL-17 in AD, we crossed mice lacking the common receptor of IL-17 signaling (IL-17RA knockout mice) to the APP/PS1 mouse model of AD. We then analyzed the composition of immune cells and cytokines/chemokines during different phases of AD pathology, and interrogated the underlying mechanism by which IL-17 may regulate immune cell infiltration into AD brains. RESULTS Ablation of IL-17RA in APP/PS1 mice decreased infiltration of CD8+ T cells and myeloid cells to mouse brain. IL-17 was able to promote the production of myeloid- and T cell-attracting chemokines CXCL1 and CXCL9/10 in primary glial cells. We also observed that IL-17 is upregulated in the late stage of AD development, and ectopic expression of IL-17 via adenoviral infection to the cortex trended towards worsened cognition in APP/PS1 mice, suggesting a pathogenic role of excessive IL-17 in AD. CONCLUSION Our data show that IL-17 signaling promotes neuroinflammation in AD by accelerating the infiltration of CD8+ T lymphocytes and Gr1+ CD11b+ myeloid cells.
Collapse
Affiliation(s)
- Xiaoyang Ye
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA.,Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Ju Chen
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jie Pan
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Qi Wu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Yue Wang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Mengqian Lu
- School of Acupuncture-moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Chengrong Zhang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Zhenzhen Zhang
- Guangxi Neurological Diseases Clinical Research Center, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Muyan Ma
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jinyong Zhu
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| | - Anthony T Vella
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jun Wan
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic & Systems Biology, TNLIST, School of Medicine, Tsinghua University, Beijing, China
| | - Kepeng Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
4
|
Rudge JD. A New Hypothesis for Alzheimer's Disease: The Lipid Invasion Model. J Alzheimers Dis Rep 2022; 6:129-161. [PMID: 35530118 PMCID: PMC9028744 DOI: 10.3233/adr-210299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/05/2022] [Indexed: 02/07/2023] Open
Abstract
This paper proposes a new hypothesis for Alzheimer's disease (AD)-the lipid invasion model. It argues that AD results from external influx of free fatty acids (FFAs) and lipid-rich lipoproteins into the brain, following disruption of the blood-brain barrier (BBB). The lipid invasion model explains how the influx of albumin-bound FFAs via a disrupted BBB induces bioenergetic changes and oxidative stress, stimulates microglia-driven neuroinflammation, and causes anterograde amnesia. It also explains how the influx of external lipoproteins, which are much larger and more lipid-rich, especially more cholesterol-rich, than those normally present in the brain, causes endosomal-lysosomal abnormalities and overproduction of the peptide amyloid-β (Aβ). This leads to the formation of amyloid plaques and neurofibrillary tangles, the most well-known hallmarks of AD. The lipid invasion model argues that a key role of the BBB is protecting the brain from external lipid access. It shows how the BBB can be damaged by excess Aβ, as well as by most other known risk factors for AD, including aging, apolipoprotein E4 (APOE4), and lifestyle factors such as hypertension, smoking, obesity, diabetes, chronic sleep deprivation, stress, and head injury. The lipid invasion model gives a new rationale for what we already know about AD, explaining its many associated risk factors and neuropathologies, including some that are less well-accounted for in other explanations of AD. It offers new insights and suggests new ways to prevent, detect, and treat this destructive disease and potentially other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonathan D’Arcy Rudge
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
5
|
Mátyás A, Borbély E, Mihály A. Hippocampal Sclerosis in Pilocarpine Epilepsy: Survival of Peptide-Containing Neurons and Learning and Memory Disturbances in the Adult NMRI Strain Mouse. Int J Mol Sci 2021; 23:ijms23010204. [PMID: 35008630 PMCID: PMC8745054 DOI: 10.3390/ijms23010204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/28/2022] Open
Abstract
The present experiments reveal the alterations of the hippocampal neuronal populations in chronic epilepsy. The mice were injected with a single dose of pilocarpine. They had status epilepticus and spontaneously recurrent motor seizures. Three months after pilocarpine treatment, the animals were investigated with the Barnes maze to determine their learning and memory capabilities. Their hippocampi were analyzed 2 weeks later (at 3.5 months) with standard immunohistochemical methods and cell counting. Every animal displayed hippocampal sclerosis. The neuronal loss was evaluated with neuronal-N immunostaining, and the activation of the microglia was measured with Iba1 immunohistochemistry. The neuropeptide Y, parvalbumin, and calretinin immunoreactive structures were qualitatively and quantitatively analyzed in the hippocampal formation. The results were compared statistically to the results of the control mice. We detected neuronal loss and strongly activated microglia populations. Neuropeptide Y was significantly upregulated in the sprouting axons. The number of parvalbumin- and calretinin-containing interneurons decreased significantly in the Ammon’s horn and dentate gyrus. The epileptic animals displayed significantly worse learning and memory functions. We concluded that degeneration of the principal neurons, a numerical decrease of PV-containing GABAergic neurons, and strong peptidergic axonal sprouting were responsible for the loss of the hippocampal learning and memory functions.
Collapse
Affiliation(s)
- Adrienne Mátyás
- Department of Anatomy, Albert Szent-Györgyi Faculty of Medicine, University of Szeged, Kossuth L. sgt. 38, H-6724 Szeged, Hungary;
| | - Emőke Borbély
- Department of Medical Chemistry, University of Szeged, Dóm tér. 8, H-6720 Szeged, Hungary;
- Professional Pedagogical Service of Csongrád-Csanád County, Űrhajós u. 4, H-6723 Szeged, Hungary
| | - András Mihály
- Department of Anatomy, Albert Szent-Györgyi Faculty of Medicine, University of Szeged, Kossuth L. sgt. 38, H-6724 Szeged, Hungary;
- Correspondence:
| |
Collapse
|
6
|
Black SAG, Stepanchuk AA, Templeton GW, Hernandez Y, Ota T, Roychoudhury S, Smith EE, Barber PA, Ismail Z, Fischer K, Zwiers A, Poulin MJ, Blennow K, Zetterberg H, Stys PK, Tsutsui S. Diagnosing Alzheimer's Disease from Circulating Blood Leukocytes Using a Fluorescent Amyloid Probe. J Alzheimers Dis 2021; 85:1721-1734. [PMID: 34958041 DOI: 10.3233/jad-215402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Toxic amyloid-β (Aβ) peptides aggregate into higher molecular weight assemblies and accumulate not only in the extracellular space, but also in the walls of blood vessels in the brain, increasing their permeability, and promoting immune cell migration and activation. Given the prominent role of the immune system, phagocytic blood cells may contact pathological brain materials. OBJECTIVE To develop a novel method for early Alzheimer's disease (AD) detection, we used blood leukocytes, that could act as "sentinels" after trafficking through the brain microvasculature, to detect pathological amyloid by labelling with a conformationally-sensitive fluorescent amyloid probe and imaging with confocal spectral microscopy. METHODS Formalin-fixed peripheral blood mononuclear cells (PBMCs) from cognitively healthy control (HC) subjects, mild cognitive impairment (MCI) and AD patients were stained with the fluorescent amyloid probe K114, and imaged. Results were validated against cerebrospinal fluid (CSF) biomarkers and clinical diagnosis. RESULTS K114-labeled leukocytes exhibited distinctive fluorescent spectral signatures in MCI/AD subjects. Comparing subjects with single CSF biomarker-positive AD/MCI to negative controls, our technique yielded modest AUCs, which improved to the 0.90 range when only MCI subjects were included in order to measure performance in an early disease state. Combining CSF Aβ 42 and t-Tau metrics further improved the AUC to 0.93. CONCLUSION Our method holds promise for sensitive detection of AD-related protein misfolding in circulating leukocytes, particularly in the early stages of disease.
Collapse
Affiliation(s)
- Stefanie A G Black
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada.,Amira Medical Technologies Inc., Calgary, AB, Canada
| | - Anastasiia A Stepanchuk
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | | | - Yda Hernandez
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Faculty of Kinesiology, University of Calgary, Calgary AB, Canada
| | - Tomoko Ota
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Shyamosree Roychoudhury
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Eric E Smith
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Philip A Barber
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada.,Calgary Stroke Program, Seaman Family MR Center, Departments of Clinical Neurosciences and Radiology, Foothills Medical Centre, Calgary AB, Canada.,Department of Community Health Sciences, University of Calgary, Calgary AB, Canada
| | - Zahinoor Ismail
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada.,Department of Psychiatry, and the Mathison Centre for Mental Health Research & Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Community Health Sciences, University of Calgary, Calgary AB, Canada.,O'Brien Institute of Public Health, University of Calgary, Calgary AB, Canada
| | - Karyn Fischer
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Angela Zwiers
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Marc J Poulin
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada.,O'Brien Institute of Public Health, University of Calgary, Calgary AB, Canada.,Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Faculty of Kinesiology, University of Calgary, Calgary AB, Canada
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, University College London Queen Square Institute of Neurology, London, UK.,UK Dementia Research Institute, University College London, London, UK
| | - Peter K Stys
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Shigeki Tsutsui
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary AB, Canada.,Amira Medical Technologies Inc., Calgary, AB, Canada
| |
Collapse
|
7
|
Uddin MS, Kabir MT, Al Mamun A, Behl T, Mansouri RA, Aloqbi AA, Perveen A, Hafeez A, Ashraf GM. Exploring Potential of Alkaloidal Phytochemicals Targeting Neuroinflammatory Signaling of Alzheimer's Disease. Curr Pharm Des 2021; 27:357-366. [PMID: 32473620 DOI: 10.2174/1381612826666200531151004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/27/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder that is marked by cognitive dysfunctions and the existence of neuropathological hallmarks such as amyloid plaques, and neurofibrillary tangles. It has been observed that a persistent immune response in the brain has appeared as another neuropathological hallmark in AD. The sustained activation of the microglia, the brain's resident macrophages, and other immune cells has been shown to aggravate both tau and amyloid pathology and may consider as a connection in the AD pathogenesis. However, the basic mechanisms that link immune responses in the pathogenesis of AD are unclear until now since the process of neuroinflammation can have either a harmful or favorable effect on AD, according to the phase of the disease. Numerous researches recommend that nutritional fruits, as well as vegetables, possess neurodefensive properties against the detrimental effects of neuroinflammation and aging. Moreover, these effects are controlled by diverse phytochemical compounds that are found in plants and demonstrate anti-inflammatory, neuroprotective, as well as other beneficial actions. In this review, we focus on the link of neuroinflammation in AD as well as highlight the probable mechanisms of alkaloidal phytochemicals to combat the neuroinflammatory aspect of AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | | | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rasha A Mansouri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
8
|
Caffeine effects on systemic metabolism, oxidative-inflammatory pathways, and exercise performance. Nutr Res 2020; 80:1-17. [DOI: 10.1016/j.nutres.2020.05.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 04/11/2020] [Accepted: 05/09/2020] [Indexed: 01/06/2023]
|
9
|
Cipollini V, Anrather J, Orzi F, Iadecola C. Th17 and Cognitive Impairment: Possible Mechanisms of Action. Front Neuroanat 2019; 13:95. [PMID: 31803028 PMCID: PMC6877481 DOI: 10.3389/fnana.2019.00095] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022] Open
Abstract
T helper 17 (Th17) cells represent a distinct population of immune cells, important in the defense of the organism against extracellular infectious agents. Because of their cytokine profile and ability to recruit other immune cell types, they are highly pro-inflammatory and are involved in the induction of several autoimmune disorders. Recent studies show that Th17 cells and their signature cytokine IL-17 have also a role in a wide variety of neurological diseases. This review article will briefly summarize the evidence linking Th17 cells to brain diseases associated with cognitive impairment, including multiple sclerosis (MS), ischemic brain injury and Alzheimer’s disease (AD). We will also investigate the mechanisms by which these cells enter the brain and induce brain damage, including direct effects of IL-17 on brain cells and indirect effects mediated through disruption of the blood-brain barrier (BBB), neurovascular dysfunction and gut-brain axis. Finally, therapeutic prospects targeting Th17 cells and IL-17 will be discussed.
Collapse
Affiliation(s)
| | - Josef Anrather
- Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Francesco Orzi
- Sant' Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Costantino Iadecola
- Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
10
|
Pan Y, Nicolazzo JA. Impact of aging, Alzheimer's disease and Parkinson's disease on the blood-brain barrier transport of therapeutics. Adv Drug Deliv Rev 2018; 135:62-74. [PMID: 29665383 DOI: 10.1016/j.addr.2018.04.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/17/2018] [Accepted: 04/07/2018] [Indexed: 01/01/2023]
Abstract
Older people are at a greater risk of medicine-induced toxicity resulting from either increased drug sensitivity or age-related pharmacokinetic changes. The scenario is further complicated with the two most prevalent age-related neurodegenerative diseases, Alzheimer's disease (AD) and Parkinson's disease (PD). With aging, AD and PD, there is growing evidence of altered structure and function of the blood-brain barrier (BBB), including modifications to tight junctions and efflux transporters, such as P-glycoprotein. The subsequent impact on CNS drug exposure and risk of neurotoxicity from systemically-acting medicines is less well characterized. The purpose of this review, therefore, is to provide an overview of the multiple changes that occur to the BBB as a result of aging, AD and PD, and the impact that such changes have on CNS exposure of drugs, based on studies conducted in aged rodents or rodent models of disease, and in elderly people with and without AD or PD.
Collapse
Affiliation(s)
- Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia.
| |
Collapse
|
11
|
Stock AJ, Kasus-Jacobi A, Pereira HA. The role of neutrophil granule proteins in neuroinflammation and Alzheimer's disease. J Neuroinflammation 2018; 15:240. [PMID: 30149799 PMCID: PMC6112130 DOI: 10.1186/s12974-018-1284-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/16/2018] [Indexed: 02/08/2023] Open
Abstract
Neutrophils are the innate immune system’s first line of defense. Neutrophils play a critical role in protecting the host against infectious pathogens, resolving sterile injuries, and mediating inflammatory responses. The granules of neutrophils and their constituent proteins are central to these functions. Although neutrophils may exert a protective role upon acute inflammatory conditions or insults, continued activity of neutrophils in chronic inflammatory diseases can contribute to tissue damage. Neutrophil granule proteins are involved in a number of chronic inflammatory conditions and diseases. However, the functions of these proteins in neuroinflammation and chronic neuroinflammatory diseases, including Alzheimer’s disease (AD), remain to be elucidated. In this review, we discuss recent findings from our lab and others that suggest possible functions for neutrophils and the neutrophil granule proteins, CAP37, neutrophil elastase, and cathepsin G, in neuroinflammation, with an emphasis on AD. These findings reveal that neutrophil granule proteins may exert both neuroprotective and neurotoxic effects. Further research should determine whether neutrophil granule proteins are valid targets for therapeutic interventions in chronic neuroinflammatory diseases.
Collapse
Affiliation(s)
- Amanda J Stock
- The Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., BRC Rm 06B121, Baltimore, MD, 21224, USA.,Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA
| | - Anne Kasus-Jacobi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA.,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA
| | - H Anne Pereira
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA. .,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA. .,Department of Cell Biology, University of Oklahoma Health Sciences Center, 1105 N. Stonewall, Robert M. Bird Library, Rm 258, Oklahoma City, OK, 73117, USA. .,Department of Pathology, University of Oklahoma Health Sciences Center, 1105 N. Stonewall, Robert M. Bird Library, Rm 258, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
12
|
A novel dynamic multicellular co-culture system for studying individual blood-brain barrier cell types in brain diseases and cytotoxicity testing. Sci Rep 2018; 8:8784. [PMID: 29884831 PMCID: PMC5993789 DOI: 10.1038/s41598-018-26480-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 05/08/2018] [Indexed: 01/10/2023] Open
Abstract
Blood brain barrier (BBB) cells play key roles in the physiology and pathology of the central nervous system (CNS). BBB dysfunction is implicated in many neurodegenerative diseases, including Alzheimer’s disease (AD). The BBB consists of capillary endothelial cells, pericytes encircling the endothelium and surrounding astrocytes extending their processes towards it. Although there have been many attempts to develop in vitro BBB models, the complex interaction between these cell types makes it extremely difficult to determine their individual contribution to neurotoxicity in vivo. Thus, we developed and optimised an in vitro multicellular co-culture model within the Kirkstall Quasi Vivo System. The main aim was to determine the optimal environment to culture human brain primary endothelial cells, pericytes and astrocytes whilst maintaining cellular communication without formation of a barrier in order to assess the contribution of each cell type to the overall response. As a proof of concept for the present system, the effects of amyloid-beta 25-35 peptide (Aβ25-35), a hallmark of AD, were explored. This multicellular system will be a valuable tool for future studies on the specific roles of individual BBB cell type (while making connection with each other through medium) in CNS disorders as well as in cytotoxicity tests.
Collapse
|
13
|
Madeira MH, Boia R, Ambrósio AF, Santiago AR. Having a Coffee Break: The Impact of Caffeine Consumption on Microglia-Mediated Inflammation in Neurodegenerative Diseases. Mediators Inflamm 2017; 2017:4761081. [PMID: 28250576 PMCID: PMC5307009 DOI: 10.1155/2017/4761081] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/12/2017] [Indexed: 12/20/2022] Open
Abstract
Caffeine is the major component of coffee and the most consumed psychostimulant in the world and at nontoxic doses acts as a nonselective adenosine receptor antagonist. Epidemiological evidence suggests that caffeine consumption reduces the risk of several neurological and neurodegenerative diseases. However, despite the beneficial effects of caffeine consumption in human health and behaviour, the mechanisms by which it impacts the pathophysiology of neurodegenerative diseases still remain to be clarified. A promising hypothesis is that caffeine controls microglia-mediated neuroinflammatory response associated with the majority of neurodegenerative conditions. Accordingly, it has been already described that the modulation of adenosine receptors, namely, the A2A receptor, affords neuroprotection through the control of microglia reactivity and neuroinflammation. In this review, we will summarize the main effects of caffeine in the modulation of neuroinflammation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria H. Madeira
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Raquel Boia
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
| | - António F. Ambrósio
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal
| | - Ana R. Santiago
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal
| |
Collapse
|
14
|
Manocha GD, Floden AM, Rausch K, Kulas JA, McGregor BA, Rojanathammanee L, Puig KR, Puig KL, Karki S, Nichols MR, Darland DC, Porter JE, Combs CK. APP Regulates Microglial Phenotype in a Mouse Model of Alzheimer's Disease. J Neurosci 2016; 36:8471-86. [PMID: 27511018 PMCID: PMC4978805 DOI: 10.1523/jneurosci.4654-15.2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 05/20/2016] [Accepted: 06/24/2016] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED Prior work suggests that amyloid precursor protein (APP) can function as a proinflammatory receptor on immune cells, such as monocytes and microglia. Therefore, we hypothesized that APP serves this function in microglia during Alzheimer's disease. Although fibrillar amyloid β (Aβ)-stimulated cytokine secretion from both wild-type and APP knock-out (mAPP(-/-)) microglial cultures, oligomeric Aβ was unable to stimulate increased secretion from mAPP(-/-) cells. This was consistent with an ability of oligomeric Aβ to bind APP. Similarly, intracerebroventricular infusions of oligomeric Aβ produced less microgliosis in mAPP(-/-) mice compared with wild-type mice. The mAPP(-/-) mice crossed to an APP/PS1 transgenic mouse line demonstrated reduced microgliosis and cytokine levels and improved memory compared with wild-type mice despite robust fibrillar Aβ plaque deposition. These data define a novel function for microglial APP in regulating their ability to acquire a proinflammatory phenotype during disease. SIGNIFICANCE STATEMENT A hallmark of Alzheimer's disease (AD) brains is the accumulation of amyloid β (Aβ) peptide within plaques robustly invested with reactive microglia. This supports the notion that Aβ stimulation of microglial activation is one source of brain inflammatory changes during disease. Aβ is a cleavage product of the ubiquitously expressed amyloid precursor protein (APP) and is able to self-associate into a wide variety of differently sized and structurally distinct multimers. In this study, we demonstrate both in vitro and in vivo that nonfibrillar, oligomeric forms of Aβ are able to interact with the parent APP protein to stimulate microglial activation. This provides a mechanism by which metabolism of APP results in possible autocrine or paracrine Aβ production to drive the microgliosis associated with AD brains.
Collapse
Affiliation(s)
- Gunjan D Manocha
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Angela M Floden
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Keiko Rausch
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Joshua A Kulas
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Brett A McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Lalida Rojanathammanee
- Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000 Thailand
| | - Kelley R Puig
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Kendra L Puig
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Sanjib Karki
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, Missouri 63121-4400, and
| | - Michael R Nichols
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, Missouri 63121-4400, and
| | - Diane C Darland
- Department of Biology, University of North Dakota, Grand Forks, North Dakota 58202
| | - James E Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Colin K Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203,
| |
Collapse
|
15
|
Di Marco LY, Venneri A, Farkas E, Evans PC, Marzo A, Frangi AF. Vascular dysfunction in the pathogenesis of Alzheimer's disease--A review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis 2015; 82:593-606. [PMID: 26311408 DOI: 10.1016/j.nbd.2015.08.014] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/23/2015] [Accepted: 08/17/2015] [Indexed: 12/17/2022] Open
Abstract
Late-onset dementia is a major health concern in the ageing population. Alzheimer's disease (AD) accounts for the largest proportion (65-70%) of dementia cases in the older population. Despite considerable research effort, the pathogenesis of late-onset AD remains unclear. Substantial evidence suggests that the neurodegenerative process is initiated by chronic cerebral hypoperfusion (CCH) caused by ageing and cardiovascular conditions. CCH causes reduced oxygen, glucose and other nutrient supply to the brain, with direct damage not only to the parenchymal cells, but also to the blood-brain barrier (BBB), a key mediator of cerebral homeostasis. BBB dysfunction mediates the indirect neurotoxic effects of CCH by promoting oxidative stress, inflammation, paracellular permeability, and dysregulation of nitric oxide, a key regulator of regional blood flow. As such, BBB dysfunction mediates a vicious circle in which cerebral perfusion is reduced further and the neurodegenerative process is accelerated. Endothelial interaction with pericytes and astrocytes could also play a role in the process. Reciprocal interactions between vascular dysfunction and neurodegeneration could further contribute to the development of the disease. A comprehensive overview of the complex scenario of interacting endothelium-mediated processes is currently lacking, and could prospectively contribute to the identification of adequate therapeutic interventions. This study reviews the current literature of in vitro and ex vivo studies on endothelium-mediated mechanisms underlying vascular dysfunction in AD pathogenesis, with the aim of presenting a comprehensive overview of the complex network of causative relationships. Particular emphasis is given to vicious circles which can accelerate the process of neurovascular degeneration.
Collapse
Affiliation(s)
- Luigi Yuri Di Marco
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK.
| | - Annalena Venneri
- Department of Neuroscience, Medical School, University of Sheffield, Sheffield, UK; IRCCS San Camillo Foundation Hospital, Venice, Italy
| | - Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Paul C Evans
- Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield, UK
| | - Alberto Marzo
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Alejandro F Frangi
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK
| |
Collapse
|
16
|
Drug Access to the Central Nervous System in Alzheimer’s Disease: Preclinical and Clinical Insights. Pharm Res 2014; 32:819-39. [DOI: 10.1007/s11095-014-1522-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/12/2014] [Indexed: 12/12/2022]
|
17
|
Zhang J, Ke KF, Liu Z, Qiu YH, Peng YP. Th17 cell-mediated neuroinflammation is involved in neurodegeneration of aβ1-42-induced Alzheimer's disease model rats. PLoS One 2013; 8:e75786. [PMID: 24124514 PMCID: PMC3790825 DOI: 10.1371/journal.pone.0075786] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 08/20/2013] [Indexed: 01/05/2023] Open
Abstract
Neuroinflammation, especially innate immunocyte-mediated neuroinflammation, has been reported to participate in pathogenesis of Alzheimer's disease (AD). However, the involvement of adaptive immune cells, such as CD4(+) T lymphocytes, in pathogenesis of AD is not well clarified. Herein, we focus on T helper 17 (Th17) cells, a subpopulation of CD4(+) T cells with high proinflammation, and show the implication of the cells in neurodegeneration of AD. Amyloid β1-42 (Aβ1-42) was bilaterally injected into hippocampus of rats to induce AD. On days 7 and 14 following the Aβ1-42 administration, escape latency of the rats in Morris water maze was increased, expression of amyloid precursor protein was upregulated, but expression of protein phosphatase 2A was downregulated in the hippocampus, and Nissl stain showed neuronal loss and gliosis in CA1 region. Infusion of FITC-linked albumin in blood circulation and combination with immunostaining of hippocampal sections for RORγ, a specific transcriptional factor of Th17 cells, demonstrated blood-brain barrier (BBB) disruption and Th17 cells' infiltration into brain parenchyma of AD rats. Expression of Th17 proinflammatory cytokines, interleukin (IL)-17 and IL-22, was increased in the hippocampus, and concentrations of the two cytokines were elevated in both the cerebrospinal fluid and the serum in AD occurrence and development. Compared with intact or saline-treated control rats, AD animals indicated an upregulated expression of Fas and FasL in the hippocampus. Further, the immunofluorescent histochemistry on AD hippocampal sections with NeuN, RORγ, Fas and FasL displayed that Fas was principally expressed by neurons and FasL was predominantly expressed by Th17 cells, and that neuronal apoptosis shown by TUNEL and NeuN double-labeled cells increased. These results suggest that Th17 cells, which were infiltrated into AD brain parenchyma, participate in neuroinflammation and neurodegeneration of AD by release of proinflammatory cytokines and by direct action on neurons via Fas/FasL apoptotic pathway.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
- Department of Neurology, Affiliated Hospital, Nantong University, Nantong, Jiangsu Province, China
| | - Kai-Fu Ke
- Department of Neurology, Affiliated Hospital, Nantong University, Nantong, Jiangsu Province, China
| | - Zhan Liu
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
18
|
Pluta R. Is the ischemic blood–brain barrier insufficiency responsible for full-blown Alzheimer's disease? Neurol Res 2013; 28:665-71. [PMID: 16945220 DOI: 10.1179/016164106x130399] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The goal of this paper is to provide scientists with a comprehensive review of the state-of-the-art influence the ischemic blood-brain barrier (BBB) has on the final development of Alzheimer's disease and to provide detailed food-for-thought which will hopefully stimulate more researchers in this area of neuroscience. Understanding new and fundamental concepts about the behavior of the BBB during long-term reperfusion after ischemia with a variety of new neuropathogenic factors can hopefully provide some interesting clues related to the pathologic processes issues that have been receiving considerable attention in the human clinic. We present the recent data to understand the role of the BBB in maturation of both diseases and try to differentiate between primary and secondary pathologic mechanisms. In conclusion, the neuropathogenesis of Alzheimer's disease involves an initial ischemic neuronal alterations leading to enhanced neuronal vulnerability to beta-amyloid peptide and the ischemic breakdown of the BBB with leakage of serum borne beta-amyloid peptide into brain parenchyma, activation of beta-amyloid peptide-dependent toxicity culminating in the formation of amyloid plaques and finally end in full-blown Alzheimer's disease. In summary, probably we have combined mechanism(s) of ischemia processes, ischemic and chronic BBB dysfunction and beta-amyloid peptide-dependent injury in pathology of neurodegeneration that is observed in Alzheimer's disease. We speculate that Alzheimer's disease may be caused by silent and sublethal ischemic episodes that attack and slowly steal the minds of its victims. Finally, our review proposes the ischemic BBB-dependent mechanism(s) that probably are responsible for full-blown Alzheimer's disease.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Neurodegenerative Disorders, Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
19
|
Drummond ES, Muhling J, Martins RN, Wijaya LK, Ehlert EM, Harvey AR. Pathology associated with AAV mediated expression of beta amyloid or C100 in adult mouse hippocampus and cerebellum. PLoS One 2013; 8:e59166. [PMID: 23516609 PMCID: PMC3596293 DOI: 10.1371/journal.pone.0059166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 02/13/2013] [Indexed: 11/18/2022] Open
Abstract
Accumulation of beta amyloid (Aβ) in the brain is a primary feature of Alzheimer’s disease (AD) but the exact molecular mechanisms by which Aβ exerts its toxic actions are not yet entirely clear. We documented pathological changes 3 and 6 months after localised injection of recombinant, bi-cistronic adeno-associated viral vectors (rAAV2) expressing human Aβ40-GFP, Aβ42-GFP, C100-GFP or C100V717F-GFP into the hippocampus and cerebellum of 8 week old male mice. Injection of all rAAV2 vectors resulted in wide-spread transduction within the hippocampus and cerebellum, as shown by expression of transgene mRNA and GFP protein. Despite the lack of accumulation of Aβ protein after injection with AAV vectors, injection of rAAV2-Aβ42-GFP and rAAV2- C100V717F-GFP into the hippocampus resulted in significantly increased microgliosis and altered permeability of the blood brain barrier, the latter revealed by high levels of immunoglobulin G (IgG) around the injection site and the presence of IgG positive cells. In comparison, injection of rAAV2-Aβ40-GFP and rAAV2-C100-GFP into the hippocampus resulted in substantially less neuropathology. Injection of rAAV2 vectors into the cerebellum resulted in similar types of pathological changes, but to a lesser degree. The use of viral vectors to express different types of Aβ and C100 is a powerful technique with which to examine the direct in vivo consequences of Aβ expression in different regions of the mature nervous system and will allow experimentation and analysis of pathological AD-like changes in a broader range of species other than mouse.
Collapse
Affiliation(s)
- Eleanor S Drummond
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Western Australia, Australia.
| | | | | | | | | | | |
Collapse
|
20
|
Weckbach S, Neher M, Losacco JT, Bolden AL, Kulik L, Flierl MA, Bell SE, Holers VM, Stahel PF. Challenging the role of adaptive immunity in neurotrauma: Rag1(-/-) mice lacking mature B and T cells do not show neuroprotection after closed head injury. J Neurotrauma 2012; 29:1233-42. [PMID: 22335783 DOI: 10.1089/neu.2011.2169] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The role of adaptive immunity in contributing to post-traumatic neuroinflammation and neuropathology after head injury remains largely unexplored. The present study was designed to investigate the pathophysiological sequelae of closed head injury in Rag1(-/-) mice devoid of mature B and T lymphocytes. C57BL/6 wild-type and Rag1(-/-) mice were subjected to experimental closed head injury, using a standardized weight-drop device. Outcome parameters consisted of neurological scoring, quantification of blood-brain barrier (BBB) function, measurement of inflammatory markers and mediators of apoptosis in serum and brain tissue, and assessment of neuronal cell death, astrogliosis, and tissue destruction. There was no difference between wild-type and Rag1(-/-) mice with regard to injury severity and neurological impairment for up to 7 days after head injury. The extent of BBB dysfunction was in a similar range for both groups. Quantification of complement activation fragments in serum revealed significantly attenuated C3a levels in Rag1(-/-) mice compared to wild-type animals. In contrast, the levels of pro- and anti-inflammatory cytokines and pro-apoptotic and anti-apoptotic mediators remained in a similar range for both groups, and the histological analysis of brain sections did not reveal a difference in reactive astrogliosis, tissue destruction, and neuronal cell death in Rag1(-/-) compared to wild-type mice. These findings suggest that adaptive immunity is not of crucial importance for initiating and sustaining the inflammatory neuropathology after closed head injury. The attenuated extent of post-traumatic complement activation seen in Rag1(-/-) mice implies a cross-talk between innate and adaptive immune responses, which requires further investigation in future studies.
Collapse
Affiliation(s)
- Sebastian Weckbach
- Department of Orthopaedics, Denver Health Medical Center, University of Colorado School of Medicine, Denver, Colorado 80204, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Alzheimer’s disease, a neurodegenerative disorder, is associated with various pathological alterations to the blood–brain barrier, including disruption to the inter-endothelial tight junction proteins, altered expression of transport proteins involved in drug efflux, a reduction in cerebral blood flow and a thickening of the brain capillary basement membrane. There are many conflicting reports on whether such changes alter the ability of endogenous proteins to extravasate into the brain parenchyma, and there are even fewer reports focusing on the potential impact of these changes on drug transport into the CNS. The purpose of this review is to critically evaluate how the reported changes to the blood–brain barrier in Alzheimer’s disease have (or have not) resulted in altered CNS drug delivery, and to highlight the requirement for more rigorous and systematic studies in this field for the benefit of drug discovery and delivery scientists.
Collapse
|
22
|
Chen X, Ghribi O, Geiger JD. Caffeine protects against disruptions of the blood-brain barrier in animal models of Alzheimer's and Parkinson's diseases. J Alzheimers Dis 2010; 20 Suppl 1:S127-41. [PMID: 20164568 DOI: 10.3233/jad-2010-1376] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sporadic Alzheimer's disease (AD) and Parkinson's disease (PD) are two of the most common neurodegenerative diseases and as such they represent major public health problems. Finding effective treatments for AD and PD represents an unmet and elusive goal largely because these diseases are chronic and progressive, and have a complicated and ill-understood pathogenesis. Although the underlying mechanisms are not fully understood, caffeine, the most commonly ingested psychoactive drug in the world, has been shown in human and animal studies to be protective against AD and PD. One mechanism implicated in the pathogenesis of AD and PD is blood-brain barrier (BBB) dysfunction and we reported recently that caffeine exerts protective effects against AD and PD at least in part by keeping the BBB intact. The present review focuses on the role of BBB dysfunction in the pathogenesis of AD and PD, caffeine's protective effects against AD and PD, and potential mechanisms whereby caffeine protects against BBB leakage.
Collapse
Affiliation(s)
- Xuesong Chen
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | | | | |
Collapse
|
23
|
Pluta R, Januszewski S, Jabłoński M, Ułamek M. Factors in Creepy Delayed Neuronal Death in Hippocampus Following Brain Ischemia–Reperfusion Injury with Long-Term Survival. BRAIN EDEMA XIV 2010; 106:37-41. [DOI: 10.1007/978-3-211-98811-4_5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
24
|
Amyloid precursor protein mediates a tyrosine kinase-dependent activation response in endothelial cells. J Neurosci 2009; 29:14451-62. [PMID: 19923279 DOI: 10.1523/jneurosci.3107-09.2009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Amyloid precursor protein (APP) is a ubiquitously expressed type 1 integral membrane protein. It has the ability to bind numerous extracellular matrix components and propagate signaling responses via its cytoplasmic phospho-tyrosine, (682)YENPTY(687), binding motif. We recently demonstrated increased protein levels of APP, phosphorylated APP (Tyr682), and beta-amyloid (Abeta) in brain vasculature of atherosclerotic and Alzheimer's disease (AD) tissue colocalizing primarily within the endothelial layer. This study demonstrates similar APP changes in peripheral vasculature from human and mouse apoE(-/-) aorta, suggesting that APP-related changes are not restricted to brain vasculature. Therefore, primary mouse aortic endothelial cells and human umbilical vein endothelial cells were used as a model system to examine the function of APP in endothelial cells. APP multimerization with an anti-N-terminal APP antibody, 22C11, to simulate ligand binding stimulated an Src kinase family-dependent increase in protein phospho-tyrosine levels, APP phosphorylation, and Abeta secretion. Furthermore, APP multimerization stimulated increased protein levels of the proinflammatory proteins, cyclooxygenase-2 and vascular cell adhesion molecule-1 also in an Src kinase family-dependent manner. Endothelial APP was also involved in mediating monocytic cell adhesion. Collectively, these data demonstrate that endothelial APP regulates immune cell adhesion and stimulates a tyrosine kinase-dependent response driving acquisition of a reactive endothelial phenotype. These APP-mediated events may serve as therapeutic targets for intervention in progressive vascular changes common to cerebrovascular disease and AD.
Collapse
|
25
|
Oomen CA, Farkas E, Roman V, van der Beek EM, Luiten PGM, Meerlo P. Resveratrol preserves cerebrovascular density and cognitive function in aging mice. Front Aging Neurosci 2009; 1:4. [PMID: 20552055 PMCID: PMC2874408 DOI: 10.3389/neuro.24.004.2009] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 11/23/2009] [Indexed: 12/14/2022] Open
Abstract
Resveratrol, a natural polyphenol abundant in grapes and red wine, has been reported to exert numerous beneficial health effects. Among others, acute neuroprotective effects of resveratrol have been described in several models of neurodegeneration, both in vitro and in vivo. In the present study we examined the neuroprotective effects of long-term dietary supplementation with resveratrol in mice on behavioral, neurochemical and cerebrovascular level. We report a preserved cognitive function in resveratrol-treated aging mice, as shown by an enhanced acquisition of a spatial Y-maze task. This was paralleled by a higher microvascular density and a lower number of microvascular abnormalities in comparison to aging non-treated control animals. We found no effects of resveratrol supplementation on cholinergic cell number or fiber density. The present findings support the hypothesis that resveratrol exerts beneficial effects on the brain by maintaining cerebrovascular health. Via this mechanism resveratrol can contribute to the preservation of cognitive function during aging.
Collapse
Affiliation(s)
- Charlotte A Oomen
- Department of Molecular Neurobiology, Center for Behavior and Neuroscience, University of Groningen Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
26
|
Nagababu E, Usatyuk PV, Enika D, Natarajan V, Rifkind JM. Vascular endothelial barrier dysfunction mediated by amyloid-beta proteins. J Alzheimers Dis 2009; 17:845-54. [PMID: 19542618 PMCID: PMC2852470 DOI: 10.3233/jad-2009-1104] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neuronal inflammation is very common in Alzheimer's disease (AD). This inflammation can be caused by infiltration of neutrophils across the blood brain barrier. Endothelial permeability changes are required for the infiltration of high molecular weight components to the brain. Deposition of toxic amyloid-beta (A beta) fibrils in the cerebral vasculature, as well as in brain neurons, has been implicated in the development of AD. This study investigates the effect of A beta fibrils on the permeability of the endothelium and the mechanism for the observed permeability changes. A beta(1-40) and A beta(1-42) fibrils, but not monomers, were found to increase permeability of bovine pulmonary arterial endothelial cells in a dose- and time dependent manner as detected by transendothelial electrical resistance. This increase in permeability is only partially (25%) inhibited by catalase and is not associated with an increase in cytosolic Ca+2 or tyrosine phosphorylation. These results indicate that hydrogen peroxide is not the primary mediator for the permeability changes. Treatment of cells with both amyloid fibrils resulted in stress fiber formation, disruption and aggregation of actin filaments, and cellular gap formation. The results of this study reveal that A beta increases the permeability of endothelium by inducing change in the cytoskeleton network.
Collapse
Affiliation(s)
- Enika Nagababu
- Molecular Dynamics Section, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224
| | - Peter V. Usatyuk
- Pulmonary and Critical Care Section, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Divya Enika
- Pulmonary and Critical Care Section, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Viswanathan Natarajan
- Pulmonary and Critical Care Section, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Joseph M. Rifkind
- Molecular Dynamics Section, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224
| |
Collapse
|
27
|
Pluta R, Amek MU. Brain ischemia and ischemic blood-brain barrier as etiological factors in sporadic Alzheimer's disease. Neuropsychiatr Dis Treat 2008; 4:855-64. [PMID: 19183778 PMCID: PMC2626921 DOI: 10.2147/ndt.s3739] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The development of neuronal death and amyloid plaques is a characteristic feature of ischemic- and Alzheimer-type dementia. An important aspect of neuronal loss and amyloid plaques are their topography and neuropathogenesis. This review was performed to present the hypothesis that different fragments of blood-borne amyloid precursor protein are able to enter the ischemic blood-brain barrier. Chronic disruption of the blood-brain barrier after ischemic injury was shown. As an effect of chronic ischemic blood-brain barrier injury, a visible connection of amyloid plaques with neurovasculature was observed. This neuropathology appears to have similar distribution and mechanisms to Alzheimer's disease. The usefulness of rival ischemic theory in elucidating the neuropathogenesis of amyloid plaques formation and neuronal death in Alzheimer's disorder is discussed.
Collapse
Affiliation(s)
- Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Department of Neurodegenerative Disorders, Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
28
|
Gonzalez-Velasquez FJ, Kotarek JA, Moss MA. Soluble aggregates of the amyloid-beta protein selectively stimulate permeability in human brain microvascular endothelial monolayers. J Neurochem 2008; 107:466-77. [PMID: 18702666 DOI: 10.1111/j.1471-4159.2008.05618.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Cerebral amyloid angiopathy associated with Alzheimer's disease is characterized by cerebrovascular deposition of the amyloid-beta protein (Abeta). Abeta elicits a number of morphological and biochemical alterations in the cerebral microvasculature, which culminate in hemorrhagic stroke. Among these changes, compromise of the blood-brain barrier has been described in Alzheimer's disease brain, transgenic animal models of Alzheimer's disease, and cell culture experiments. In the current study, presented data illustrates that isolated soluble Abeta(1-40) aggregates, but not unaggregated monomer or mature fibril, enhance permeability in human brain microvascular endothelial monolayers. Abeta(1-40)-induced changes in permeability are paralleled by both a decrease in transendothelial electrical resistance and a re-localization of the tight junction-associated protein zonula occludin-1 away from cell borders and into the cytoplasm. Small soluble Abeta(1-40) aggregates are confirmed to be the most potent stimulators of endothelial monolayer permeability by establishing an inverse relationship between average aggregate size and stimulated changes in diffusional permeability coefficients. These results support previous findings demonstrating that small soluble Abeta(1-40) aggregates are also primarily responsible for endothelial activation, suggesting that these same species may elicit other changes in the cerebrovasculature associated with cerebral amyloid angiopathy and Alzheimer's disease.
Collapse
Affiliation(s)
- Francisco J Gonzalez-Velasquez
- Department of Chemical Engineering, University of South Carolina, Swearingen Engineering Center, Columbia, South Carolina, USA
| | | | | |
Collapse
|
29
|
Mangano EN, Hayley S. Inflammatory priming of the substantia nigra influences the impact of later paraquat exposure: Neuroimmune sensitization of neurodegeneration. Neurobiol Aging 2008; 30:1361-78. [PMID: 18187236 DOI: 10.1016/j.neurobiolaging.2007.11.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Revised: 10/04/2007] [Accepted: 11/16/2007] [Indexed: 12/21/2022]
Abstract
Activation of microglia along with the release of inflammatory cytokines and oxidative factors often accompanies toxin-induced degeneration of substantia nigra pars compacta (SNc) dopamine (DA) neurons. Multiple toxin exposure may synergistically influence microglial-dependent DA neuronal loss and, in fact, pre-treatment with one toxin may sensitize DA neurons to the impact of subsequent insults. Thus, we assessed whether priming SNc neurons with the inflammatory agent, lipopolysaccharide (LPS), influenced the impact of later exposure to the pesticide, paraquat, which has been reported to provoke DA loss. Indeed, LPS infusion into the SNc sensitized DA neurons to the neurodegenerative effects of a series of paraquat injections commencing 2 days later. In contrast, LPS pre-treatment actually protected against some of neurodegenerative effects of paraquat when the pesticide was administered 7 days after the endotoxin. These sensitization and de-sensitization effects were associated with altered expression of reactive microglia expressing inducible immunoproteasome subunits, as well as variations of fibroblast growth factor and a time-dependent infiltration of peripheral immune cells. Circulating levels of the inflammatory cytokines, interleukin (IL)-6, IL-2, tumor necrosis factor-alpha and interferon-gamma were also time-dependently elevated following intra-SNc LPS infusion. These data suggest that inflammatory priming may influence DA neuronal sensitivity to subsequent environmental toxins by modulating the state of glial and immune factors, and these findings may be important for neurodegenerative conditions, such as Parkinson's disease (PD).
Collapse
Affiliation(s)
- Emily N Mangano
- Institute of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | | |
Collapse
|
30
|
Arshavsky YI. Alzheimer's disease, brain immune privilege and memory: a hypothesis. J Neural Transm (Vienna) 2006; 113:1697-707. [PMID: 16932992 DOI: 10.1007/s00702-006-0524-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Accepted: 04/28/2006] [Indexed: 12/13/2022]
Abstract
The most distinctive feature of Alzheimer's disease (AD) is the specific degeneration of the neurons involved in memory consolidation, storage, and retrieval. Patients suffering from AD forget basic information about their past, loose linguistic and calculative abilities and communication skills. Thus, understanding the etiology of AD may provide insights into the mechanisms of memory and vice versa. The brain is an immunologically privileged site protected from the organism's immune reactions by the blood-brain barrier (BBB). All risk factors for AD (both cardiovascular and genetic) lead to destruction of the BBB. Evidence emerging from recent literature indicates that AD may have an autoimmune nature associated with BBB impairments. This hypothesis suggests that the process of memory consolidation involves the synthesis of novel macromolecules recognized by the immune system as "non-self" antigens. The objective of this paper is to stimulate new approaches to studies of neural mechanisms underpinning memory consolidation and its breakdown during AD. If the hypothesis on the autoimmune nature of AD is correct, the identification of the putative antigenic macromolecules might be critical to understanding the etiology and prevention of AD, as well as for elucidating cellular mechanisms of learning and memory.
Collapse
Affiliation(s)
- Y I Arshavsky
- Institute for Nonlinear Science, University of California San Diego, La Jolla, CA 92093-0402, USA.
| |
Collapse
|
31
|
Farkas E, Süle Z, Tóth-Szuki V, Mátyás A, Antal P, Farkas IG, Mihály A, Bari F. Tumor necrosis factor-alpha increases cerebral blood flow and ultrastructural capillary damage through the release of nitric oxide in the rat brain. Microvasc Res 2006; 72:113-9. [PMID: 16854437 DOI: 10.1016/j.mvr.2006.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2006] [Revised: 05/18/2006] [Accepted: 05/18/2006] [Indexed: 11/20/2022]
Abstract
Tumor necrosis factor-alpha (TNFalpha) is a proinflammatory cytokine implicated in cerebrovascular pathology. The aim of the present study was to characterize the simultaneous effects of an intracarotid administration of TNFalpha on cerebral blood flow (CBF) and the ultrastructure of the blood-brain barrier (BBB) and to determine whether nitric oxide (NO) is a mediator of the TNFalpha-induced alterations in CBF and BBB. TNFalpha (2.5 microg/kg) or saline was infused into the right common carotid artery of male Wistar rats (n = 70). NO production was inhibited with L-NAME (20 mg/kg, i.v.). CBF was monitored for 2 h with laser-Doppler flowmetry. Tissue samples were taken from the unilateral frontoparietal cortex and prepared for electron microscopy. The proportion of capillaries with swollen astrocytic endfeet and the lumen diameter of the capillaries were measured. TNFalpha significantly increased CBF, which reached a maximum of 190% of the baseline 1 h after the cessation of TNFalpha infusion. L-NAME completely prevented the increase in CBF. TNFalpha elevated the swelling of the astrocytic endfeet from a baseline value of 22.4 +/- 9.35% to 64.9 +/- 3.16%. The administration of L-NAME before TNFalpha infusion prevented the astrocytic swelling. These results demonstrate that TNFalpha increases CBF and the swelling of astrocytes through the production of NO. Our data additionally demonstrate that the breakdown of the BBB by circulating TNFalpha may involve the astrocytic endfeet.
Collapse
Affiliation(s)
- Eszter Farkas
- Department of Anatomy, School of Medicine, University of Szeged, Szeged, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Carvey PM, Zhao CH, Hendey B, Lum H, Trachtenberg J, Desai BS, Snyder J, Zhu YG, Ling ZD. 6-Hydroxydopamine-induced alterations in blood-brain barrier permeability. Eur J Neurosci 2006; 22:1158-68. [PMID: 16176358 DOI: 10.1111/j.1460-9568.2005.04281.x] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Vascular inflammation is well known for its ability to compromise the function of the blood--brain barrier (BBB). Whether inflammation on the parenchymal side of the barrier, such as that associated with Parkinson's-like dopamine (DA) neuron lesions, similarly disrupts BBB function, is unknown. We assessed BBB integrity by examining the leakage of FITC-labeled albumin or horseradish peroxidase from the vasculature into parenchyma in animals exposed to the DA neurotoxin 6-hydroxydopamine (6OHDA). Unilateral injections of 6OHDA into the striatum or the medial forebrain bundle produced increased leakage in the ipsilateral substantia nigra and striatum 10 and 34 days following 6OHDA. Microglia were markedly activated and DA neurons were reduced by the lesions. The areas of BBB leakage were associated with increased expression of P-glycoprotein and beta 3-integrin expression suggesting, respectively, a compensatory response to inflammation and possible angiogenesis. Behavioural studies revealed that domperidone, a DA antagonist that normally does not cross the BBB, attenuated apomorphine-induced stereotypic behaviour in animals with 6OHDA lesions. This suggests that drugs which normally have no effect in brain can enter following Parkinson-like lesions. These data suggest that the events associated with DA neuron loss compromise BBB function.
Collapse
Affiliation(s)
- P M Carvey
- Rush University Medical Center, Department of Pharmacology, Cohn 406, Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Huber JD, Campos CR, Mark KS, Davis TP. Alterations in blood-brain barrier ICAM-1 expression and brain microglial activation after lambda-carrageenan-induced inflammatory pain. Am J Physiol Heart Circ Physiol 2005; 290:H732-40. [PMID: 16199477 PMCID: PMC3915803 DOI: 10.1152/ajpheart.00747.2005] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies showed that peripheral inflammatory pain increased blood-brain barrier (BBB) permeability and altered tight junction protein expression and the delivery of opioid analgesics to the brain. What remains unknown is which pathways and mediators during peripheral inflammation affect BBB function and structure. The current study investigated effects of lambda-carrageenan-induced inflammatory pain (CIP) on BBB expression of ICAM-1. We also examined the systemic contribution of a number of proinflammatory cytokines and microglial activation in the brain to elucidate pathways involved in BBB disruption during CIP. We investigated ICAM-1 RNA and protein expression levels in isolated rat brain microvessels after CIP using RT-PCR and Western blot analyses, screened inflammatory cytokines during the time course of inflammation, assessed white blood cell counts, and probed for BBB and central nervous system stimulation and leukocyte transmigration using immunohistochemistry and flow cytometry. Results showed an early increase in ICAM-1 RNA and protein expression after CIP with no change in circulating levels of several proinflammatory cytokines. Changes in ICAM-1 protein expression were noted at 48 h. Immunohistochemistry showed that the induction of ICAM-1 was region specific with increased expression noted in the thalamus and frontal and parietal cortices, which directly correlated with increased expression of activated microglia. The findings of the present study were that CIP induces increased ICAM-1 mRNA and protein expression at the BBB and that systemic proinflammatory mediators play no apparent role in the early response (1-6 h); however, brain region-specific increases in microglial activation suggest a potential for a central-mediated response.
Collapse
Affiliation(s)
- J. D. Huber
- Department of Basic Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia
| | - C. R. Campos
- Department of Medical Pharmacology, University of Arizona, Tucson, Arizona
| | - K. S. Mark
- Department of Pharmacology, University of Missouri-Kansas City, Kansas City, Missouri
| | - T. P. Davis
- Department of Medical Pharmacology, University of Arizona, Tucson, Arizona
| |
Collapse
|
34
|
Besser MJ, Ganor Y, Levite M. Dopamine by itself activates either D2, D3 or D1/D5 dopaminergic receptors in normal human T-cells and triggers the selective secretion of either IL-10, TNFalpha or both. J Neuroimmunol 2005; 169:161-71. [PMID: 16150496 DOI: 10.1016/j.jneuroim.2005.07.013] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2005] [Accepted: 07/25/2005] [Indexed: 11/18/2022]
Abstract
The neurotransmitter dopamine on its own increased significantly TNFalpha and IL-10 secretion by resting normal-human T-cells, and induced approximately 5-fold elevation of the corresponding mRNA's, without affecting IFNgamma and IL-4. Using seven highly selective dopamine-receptor (DR) agonists and antagonists we found that TNFalpha-upregulation, evident after 24 h, was mediated by D3R and D1/D5R while IL-10-upregulation, evident after 72 h, was mediated by D2R and D1/D5R. We confirmed the expression of D2R and D3R in these human T cells. Dopamine's unique ability to trigger a selective secretion of either TNFalpha only (via D3R) or IL-10 only (via D2R) or both (via D1/D5R), differs from the robust and non-selective cytokine-secretion induced by 'classical' TCR-activation, and as such may have important beneficial or detrimental implications in various immunological and neurological diseases/injuries/cancers.
Collapse
Affiliation(s)
- Michal J Besser
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
| | | | | |
Collapse
|
35
|
Fabene PF, Weiczner R, Marzola P, Nicolato E, Calderan L, Andrioli A, Farkas E, Süle Z, Mihaly A, Sbarbati A. Structural and functional MRI following 4-aminopyridine-induced seizures: a comparative imaging and anatomical study. Neurobiol Dis 2005; 21:80-9. [PMID: 16084733 DOI: 10.1016/j.nbd.2005.06.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2005] [Revised: 06/11/2005] [Accepted: 06/16/2005] [Indexed: 11/25/2022] Open
Abstract
Structural and functional MRI was used in conjunction with computerized electron microscopy morphometry to study changes 2 h, 24 h and 3 days after 4-aminopyridine-induced seizures lasting 2 h in rats. T2 (relaxation time) values showed changes throughout the cerebral cortex, hippocampus, amygdala and medial thalamus, with a different temporal progression, showing a complete recovery only after 3 days. Two hours after seizures, the apparent diffusion coefficient was decreased throughout the brain compared to control animals, and a further decrease was evident 24 h after seizures. This was followed by a complete recovery at 3 days post-seizures. Functional MRI was performed using regional cerebral blood volume (rCBV) maps. The rCBV was increased shortly after convulsions (2 h) in all structures investigated, with a significant return to baseline values in the parietal cortex and hippocampus, but not in the medial thalamic nuclei, 24 h after seizure onset. No rCBV alterations were detected 3 days after seizures. Electron microscopy of tissue samples of parietal neocortex and hippocampus revealed prominent astrocytic swelling 2 h post-convulsions which decreased thereafter gradually. In conclusion, this experiment reports for the first time structural and functional brain alterations, lasting several hours, in 4-aminopyridine-treated rats after seizure onset. MRI approach combined with histological and ultrastructural analysis provided a clarification of the mechanisms involved in the brain acute response to ictal activity.
Collapse
Affiliation(s)
- P F Fabene
- Section of Anatomy and Histology, Department of Morphological and Biomedical Sciences, Faculty of Medicine, Strada Le Grazie 8, 37134 Verona, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|