151
|
Influence of microglia and astrocyte activation in the neuroinflammatory pathogenesis of Alzheimer’s disease: Rational insights for the therapeutic approaches. J Clin Neurosci 2019; 59:6-11. [DOI: 10.1016/j.jocn.2018.10.034] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/05/2018] [Indexed: 01/08/2023]
|
152
|
Cong L, Zhao Y, Pogue AI, Lukiw WJ. Role of microRNA (miRNA) and Viroids in Lethal Diseases of Plants and Animals. Potential Contribution to Human Neurodegenerative Disorders. BIOCHEMISTRY (MOSCOW) 2018; 83:1018-1029. [PMID: 30472940 DOI: 10.1134/s0006297918090031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Both plants and animals have adopted a common strategy of using ~18-25-nucleotide small non-coding RNAs (sncRNAs), known as microRNAs (miRNAs), to transmit DNA-based epigenetic information. miRNAs (i) shape the total transcriptional output of individual cells; (ii) regulate and fine-tune gene expression profiles of cell clusters, and (iii) modulate cell phenotype in response to environmental stimuli and stressors. These miRNAs, the smallest known carriers of gene-encoded post-transcriptional regulatory information, not only regulate cellular function in healthy cells but also act as important mediators in the development of plant and animal diseases. Plants possess their own specific miRNAs; at least 32 plant species have been found to carry infectious sncRNAs called viroids, whose mechanisms of generation and functions are strikingly similar to those of miRNAs. This review highlights recent remarkable and sometimes controversial findings in miRNA signaling in plants and animals. Special attention is given to the intriguing possibility that dietary miRNAs and/or sncRNAs can function as mobile epigenetic and/or evolutionary linkers between different species and contribute to both intra- and interkingdom signaling. Wherever possible, emphasis has been placed on the relevance of these miRNAs to the development of human neurodegenerative diseases, such as Alzheimer's disease. Based on the current available data, we suggest that such xeno-miRNAs may (i) contribute to the beneficial properties of medicinal plants, (ii) contribute to the negative properties of disease-causing or poisonous plants, and (iii) provide cross-species communication between kingdoms of living organisms involving multiple epigenetic and/or potentially pathogenic mechanisms associated with the onset and pathogenesis of various diseases.
Collapse
Affiliation(s)
- L Cong
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112-2272, USA.,Department of Neurology, Shengjing Hospital, China Medical University, Heping District, Shenyang, Liaoning Province, China
| | - Y Zhao
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112-2272, USA.,Department of Anatomy and Cell Biology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112-2272, USA
| | - A I Pogue
- Alchem Biotech Research, Toronto, ON M5S 1A8, Canada
| | - W J Lukiw
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112-2272, USA. .,Department Neurology, Louisiana State University School of Medicine, New Orleans, LA 70112-2272, USA.,Department Ophthalmology, Louisiana State University School of Medicine, New Orleans, LA 70112-2272, USA
| |
Collapse
|
153
|
Lukiw WJ, Cong L, Jaber V, Zhao Y. Microbiome-Derived Lipopolysaccharide (LPS) Selectively Inhibits Neurofilament Light Chain (NF-L) Gene Expression in Human Neuronal-Glial (HNG) Cells in Primary Culture. Front Neurosci 2018; 12:896. [PMID: 30568571 PMCID: PMC6289986 DOI: 10.3389/fnins.2018.00896] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 11/16/2018] [Indexed: 01/30/2023] Open
Abstract
The remarkable co-localization of highly pro-inflammatory lipopolysaccharide (LPS) with sporadic Alzheimer's disease (AD)-affected neuronal nuclei suggests that there may be some novel pathogenic contribution of this heat stable neurotoxin to neuronal activity and neuron-specific gene expression. In this communication we show for the first time: (i) the association and envelopment of sporadic AD neuronal nuclei with LPS in multiple AD neocortical tissue samples; and (ii) a selective repression in the output of neuron-specific neurofilament light (NF-L) chain messenger RNA (mRNA), perhaps as a consequence of this association. The down-regulation of NF-L mRNA and protein is a characteristic attribute of AD brain and accompanies neuronal atrophy and an associated loss of neuronal architecture with synaptic deficits. To study this phenomenon further, human neuronal-glial (HNG) cells in primary culture were incubated with LPS, and DNA arrays, Northern, Western, and ELISA analyses were used to quantify transcription patterns for the three member neuron-specific intermediate filament-gene family NF-H, NF-M, and NF-L. As in sporadic AD limbic-regions, down-regulated transcription products for the NF-L intermediate filament protein was significant. These results support our novel hypothesis: (i) that internally sourced, microbiome-derived neurotoxins such as LPS contribute to a progressive disruption in the read-out of neuron-specific genetic-information; (ii) that the presence of LPS-enveloped neuronal nuclei is associated with a down-regulation in NF-L expression, a key neuron-specific cytoskeletal component; and (iii) this may have a bearing on progressive neuronal atrophy, loss of synaptic-contact and disruption of neuronal architecture, all of which are characteristic pathological features of sporadic-AD brain. This is the first report that provides evidence for a neuron-specific effect of a human GI-tract microbiome-derived neurotoxin on decreased NF-L abundance in both sporadic AD temporal lobe neocortex in vivo and in LPS-stressed HNG cells in vitro.
Collapse
Affiliation(s)
- Walter J. Lukiw
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Department of Neurology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Department of Ophthalmology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Lin Cong
- Department of Neurology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Vivian Jaber
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Yuhai Zhao
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Department of Anatomy and Cell Biology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
154
|
Giau VV, Wu SY, Jamerlan A, An SSA, Kim SY, Hulme J. Gut Microbiota and Their Neuroinflammatory Implications in Alzheimer's Disease. Nutrients 2018; 10:nu10111765. [PMID: 30441866 PMCID: PMC6266223 DOI: 10.3390/nu10111765] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/08/2018] [Accepted: 11/10/2018] [Indexed: 12/12/2022] Open
Abstract
The bidirectional communication between the central nervous system (CNS) and the gut microbiota plays a pivotal role in human health. Increasing numbers of studies suggest that the gut microbiota can influence the brain and behavior of patients. Various metabolites secreted by the gut microbiota can affect the cognitive ability of patients diagnosed with neurodegenerative diseases. Nearly one in every ten Korean senior citizens suffers from Alzheimer’s disease (AD), the most common form of dementia. This review highlights the impact of metabolites from the gut microbiota on communication pathways between the brain and gut, as well as the neuroinflammatory roles they may have in AD patients. The objectives of this review are as follows: (1) to examine the role of the intestinal microbiota in homeostatic communication between the gut microbiota and the brain, termed the microbiota–gut–brain (MGB) axis; (2) to determine the underlying mechanisms of signal dysfunction; and (3) to assess the impact of signal dysfunction induced by the microbiota on AD. This review will aid in understanding the microbiota of elderly people and the neuroinflammatory roles they may have in AD.
Collapse
Affiliation(s)
- Vo Van Giau
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Si Ying Wu
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Angelo Jamerlan
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Seong Soo A An
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Sang Yun Kim
- Department of Neurology, Seoul National University College of Medicine & Neurocognitive Behavior Center, Seoul National University Bundang Hospital, Seoul 100-011, Korea.
| | - John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| |
Collapse
|
155
|
Anghelescu IG, Edwards D, Seifritz E, Kasper S. Stress management and the role of Rhodiola rosea: a review. Int J Psychiatry Clin Pract 2018; 22:242-252. [PMID: 29325481 DOI: 10.1080/13651501.2017.1417442] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Objective: Stress describes the physiological reaction to threat or pressure, which manifests as physical symptoms of exhaustion or energy loss and psychological symptoms, including irritability or tension. If untreated, chronic stress or burnout may develop, both are areas of unmet medical need. Evidence-based treatment and prevention measures are needed. Methods: Prevention strategies and existing treatment options for stress-related symptoms were evaluated to establish criteria for an adequate pharmacological approach to stress. The authors reviewed the literature to reach a clinically meaningful strategy for prevention and treatment of persistent stress symptoms and their consequences, including burnout and secondary diseases. Results: Current medication reveals a treatment gap. Most drugs target only psychological or physical stress symptoms. Furthermore, psychotropic medications sometimes prescribed for stress often have unacceptable side effects and bear a risk of overtreatment. Ideally pharmacological therapy should afford comprehensive treatment of all stress symptoms with a favourable safety profile. Conclusions: Rhodiola rosea extract (RRE) fulfils important requirements. It is the main adaptogen approved by the HMPC/EMA for the indication 'stress' and influences the release of stress hormones while boosting energy metabolism as revealed in animal literature. RRE offers comprehensive treatment of stress symptoms and can prevent chronic stress and stress-related complications.
Collapse
Affiliation(s)
- Ion-George Anghelescu
- a Department of Psychiatry and Psychotherapy , Clinic Dr. Fontheim , Liebenburg , Germany
| | | | - Erich Seifritz
- c Department of Psychiatry, Psychotherapy and Psychosomatics , Psychiatric Hospital, University of Zurich , Zurich , Switzerland
| | - Siegfried Kasper
- d Department of Psychiatry and Psychotherapy , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
156
|
Houghtaling J, List J, Mayer M. Nanopore-Based, Rapid Characterization of Individual Amyloid Particles in Solution: Concepts, Challenges, and Prospects. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802412. [PMID: 30225962 DOI: 10.1002/smll.201802412] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/15/2018] [Indexed: 06/08/2023]
Abstract
Aggregates of misfolded proteins are associated with several devastating neurodegenerative diseases. These so-called amyloids are therefore explored as biomarkers for the diagnosis of dementia and other disorders, as well as for monitoring disease progression and assessment of the efficacy of therapeutic interventions. Quantification and characterization of amyloids as biomarkers is particularly demanding because the same amyloid-forming protein can exist in different states of assembly, ranging from nanometer-sized monomers to micrometer-long fibrils that interchange dynamically both in vivo and in samples from body fluids ex vivo. Soluble oligomeric amyloid aggregates, in particular, are associated with neurotoxic effects, and their molecular organization, size, and shape appear to determine their toxicity. This concept article proposes that the emerging field of nanopore-based analytics on a single molecule and single aggregate level holds the potential to account for the heterogeneity of amyloid samples and to characterize these particles-rapidly, label-free, and in aqueous solution-with regard to their size, shape, and abundance. The article describes the concept of nanopore-based resistive pulse sensing, reviews previous work in amyloid analysis, and discusses limitations and challenges that will need to be overcome to realize the full potential of amyloid characterization on a single-particle level.
Collapse
Affiliation(s)
- Jared Houghtaling
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Jonathan List
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Michael Mayer
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| |
Collapse
|
157
|
Sollini M, Berchiolli R, Kirienko M, Rossi A, Glaudemans AWJM, Slart R, Erba PA. PET/MRI in Infection and Inflammation. Semin Nucl Med 2018; 48:225-241. [PMID: 29626940 DOI: 10.1053/j.semnuclmed.2018.02.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hybrid positron emission tomography/magnetic resonance imaging (PET/MR) systems are now more and more available for clinical use. PET/MR combines the unique features of MR including excellent soft tissue contrast, diffusion-weighted imaging, dynamic contrast-enhanced imaging, fMRI and other specialized sequences as well as MR spectroscopy with the quantitative physiologic information that is provided by PET. Most of the evidence of the potential clinical utility of PET/MRI is available for neuroimaging. Other areas, where PET/MR can play a larger role include head and neck, upper abdominal, and pelvic tumours. Although the role of PET/MR in infection and inflammation of the cardiovascular system and in musculoskeletal applications are promising, these areas of clinical investigation are still in the early phase and it may be a little longer before these areas reach their full potential in clinical practice. In this review, we outline the potential of hybrid PET/MR for imaging infection and inflammation. A background to the main radiopharmaceuticals and some technical considerations are also included.
Collapse
Affiliation(s)
- Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Raffaella Berchiolli
- Vascular Surgery Unit Department of Translational Research and Advanced Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Margarita Kirienko
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Alexia Rossi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - A W J M Glaudemans
- University of Groningen, University Medical Center Groningen, Medical Imaging Center, Groningen, The Netherlands
| | - Riemer Slart
- University of Groningen, University Medical Center Groningen, Medical Imaging Center, Groningen, The Netherlands.; University of Twente, Faculty of Science and Technology, Biomedical Photonic Imaging, Enschede, The Netherlands
| | - Paola Anna Erba
- Regional Center of Nuclear Medicine, Department of Translational Research and Advanced, Technologies in Medicine, University of Pisa, Pisa, Italy..
| |
Collapse
|
158
|
Patel V, Saxena S, Lund C, Thornicroft G, Baingana F, Bolton P, Chisholm D, Collins PY, Cooper JL, Eaton J, Herrman H, Herzallah MM, Huang Y, Jordans MJD, Kleinman A, Medina-Mora ME, Morgan E, Niaz U, Omigbodun O, Prince M, Rahman A, Saraceno B, Sarkar BK, De Silva M, Singh I, Stein DJ, Sunkel C, UnÜtzer JÜ. The Lancet Commission on global mental health and sustainable development. Lancet 2018; 392:1553-1598. [PMID: 30314863 DOI: 10.1016/s0140-6736(18)31612-x] [Citation(s) in RCA: 1369] [Impact Index Per Article: 195.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 06/11/2018] [Accepted: 07/05/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Vikram Patel
- Harvard Medical School, Boston, MA, USA; Harvard TH Chan School of Public Health, Boston, MA, USA; Sangath, Goa, India; Public Health Foundation of India, New Delhi, India.
| | - Shekhar Saxena
- Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Crick Lund
- Centre for Global Mental Health, Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Alan J Flisher Centre for Public Mental Health, Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Graham Thornicroft
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London UK
| | - Florence Baingana
- WHO Sierra Leone, Freetown, Sierra Leone; Makerere University School of Public Health, Kampala, Uganda
| | - Paul Bolton
- Department of International Health and Department of Mental Health, Center for Humanitarian Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Dan Chisholm
- WHO Regional Office for Europe, Copenhagen, Denmark
| | - Pamela Y Collins
- University of Washington School of Medicine and School of Public Health, Seattle, WA, USA
| | - Janice L Cooper
- The Carter Center, Monrovia, Liberia; Emory University, Atlanta, GA, USA
| | - Julian Eaton
- CBM International, Bensheim, Germany; Centre for Global Mental Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Helen Herrman
- Orygen, National Centre of Excellence in Youth Mental Health and Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia; World Psychiatric Association, Melbourne, VIC Australia; WHO Collaborating Centre in Mental Health, Melbourne, VIC Australia
| | - Mohammad M Herzallah
- Palestinian Neuroscience Initiative, Al-Quds University, Jerusalem, Palestine; Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ, USA
| | - Yueqin Huang
- National Clinical Research Centre for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Mark J D Jordans
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK; Research and Development, War Child, Amsterdam, Netherlands; Faculty of Social and Behavioural Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Arthur Kleinman
- Department of Anthropology, Harvard University, Cambridge, MA, USA; Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Ellen Morgan
- Templeton World Charity Foundation, Nassau, The Bahamas
| | - Unaiza Niaz
- Psychiatric Clinic and Stress Research Centre, Karachi, Pakistan; University of Health Sciences, Lahore, Pakistan; Dow University of Health Sciences, Karachi, Pakistan
| | - Olayinka Omigbodun
- College of Medicine and Centre for Child and Adolescent Mental Health, University of Ibadan, Ibadan, Nigeria
| | - Martin Prince
- King's Global Health Institute, King's College London, London, UK
| | - Atif Rahman
- University of Liverpool, Liverpool, UK; Human Development Research Foundation, Islamabad, Pakistan
| | - Benedetto Saraceno
- School of Medical Sciences, University Nova of Lisbon, Lisbon, Portugal; Lisbon Institute of Global Mental Health, Lisbon, Portugal
| | - Bidyut K Sarkar
- PRIDE Project, Sangath, India; Public Health Foundation of India, New Delhi, India
| | | | - Ilina Singh
- Department of Psychiatry and Wellcome Centre for Ethics and Humanities, University of Oxford, Oxford, UK
| | - Dan J Stein
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa; Groote Schuur Hospital, Cape Town, South Africa; South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Cape Town, South Africa
| | - Charlene Sunkel
- South African Federation for Mental Health, Johannesburg, South Africa; Movement for Global Mental Health, Johannesburg, South Africa
| | - JÜrgen UnÜtzer
- Department of Psychiatry and Behavioral Sciences and the Advancing Integrated Mental Health Solutions Center, University of Washington, Seattle, WA, USA
| |
Collapse
|
159
|
Physical exercise reserved amyloid-beta induced brain dysfunctions by regulating hippocampal neurogenesis and inflammatory response via MAPK signaling. Brain Res 2018; 1697:1-9. [DOI: 10.1016/j.brainres.2018.04.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/28/2018] [Accepted: 04/30/2018] [Indexed: 12/22/2022]
|
160
|
Kim HJ, Jung SW, Kim SY, Cho IH, Kim HC, Rhim H, Kim M, Nah SY. Panax ginseng as an adjuvant treatment for Alzheimer's disease. J Ginseng Res 2018; 42:401-411. [PMID: 30337800 PMCID: PMC6190533 DOI: 10.1016/j.jgr.2017.12.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 11/29/2017] [Accepted: 12/12/2017] [Indexed: 01/22/2023] Open
Abstract
Longevity in medicine can be defined as a long life without mental or physical deficits. This can be prevented by Alzheimer's disease (AD). Current conventional AD treatments only alleviate the symptoms without reversing AD progression. Recent studies demonstrated that Panax ginseng extract improves AD symptoms in patients with AD, and the two main components of ginseng might contribute to AD amelioration. Ginsenosides show various AD-related neuroprotective effects. Gintonin is a newly identified ginseng constituent that contains lysophosphatidic acids and attenuates AD-related brain neuropathies. Ginsenosides decrease amyloid β-protein (Aβ) formation by inhibiting β- and γ-secretase activity or by activating the nonamyloidogenic pathway, inhibit acetylcholinesterase activity and Aβ-induced neurotoxicity, and decrease Aβ-induced production of reactive oxygen species and neuroinflammatory reactions. Oral administration of ginsenosides increases the expression levels of enzymes involved in acetylcholine synthesis in the brain and alleviates Aβ-induced cholinergic deficits in AD models. Similarly, gintonin inhibits Aβ-induced neurotoxicity and activates the nonamyloidogenic pathway to reduce Aβ formation and to increase acetylcholine and choline acetyltransferase expression in the brain through lysophosphatidic acid receptors. Oral administration of gintonin attenuates brain amyloid plaque deposits, boosting hippocampal cholinergic systems and neurogenesis, thereby ameliorating learning and memory impairments. It also improves cognitive functions in patients with AD. Ginsenosides and gintonin attenuate AD-related neuropathology through multiple routes. This review focuses research demonstrating that ginseng constituents could be a candidate as an adjuvant for AD treatment. However, clinical investigations including efficacy and tolerability analyses may be necessary for the clinical acceptance of ginseng components in combination with conventional AD drugs.
Collapse
Key Words
- AChE, acetylcholinesterase
- AD, Alzheimer's disease
- APP, amyloid precursor protein
- Adjuvant
- Alzheimer's disease
- Aβ, amyloid β-protein
- BDNF, brain-derived neurotrophic factor
- EGF, Epidermal growth factor
- GLP151, ginseng major latex-like protein 151
- Ginsenoside
- Gintonin
- LPA, Lysophosphatidic acid
- NGF, nerve growth factor
- NMDA, n-methyl-d-aspartic acid
- PI3K, phosphoinositide-3 kinase
- PPARγ, peroxisome proliferator-activated receptor-γ
- Panax ginseng
- ROS, reactive oxygen species
- sAPPα, soluble amyloid precursor protein α
Collapse
Affiliation(s)
- Hyeon-Joong Kim
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Seok-Won Jung
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Seog-Young Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine and Institute of Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and toxicology program, College of Pharmacy, Kangwon National University, Chunchon, Republic of Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Manho Kim
- Department of Neurology, Neuroscience Research Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
161
|
ATP/P2X7 receptor signaling as a potential anti-inflammatory target of natural polyphenols. PLoS One 2018; 13:e0204229. [PMID: 30248132 PMCID: PMC6152980 DOI: 10.1371/journal.pone.0204229] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 09/05/2018] [Indexed: 12/13/2022] Open
Abstract
Innate immune cells, such as macrophages, respond to pathogen-associated molecular patterns, such as a lipopolysaccharide (LPS), to secrete various inflammatory mediators. Recent studies have suggested that damage-associated molecular patterns (DAMPs), released extracellularly from damaged or immune cells, also play a role in the activation of inflammatory responses. In this study, to prevent excess inflammation, we focused on DAMPs-mediated signaling that promotes LPS-stimulated inflammatory responses, especially adenosine 5’-triphosphate (ATP)-triggered signaling through the ionotropic purinergic receptor 7 (P2X7R), as a potential new anti-inflammatory target of natural polyphenols. We focused on the phenomenon that ATP accelerates the production of inflammatory mediators, such as nitric oxide, in LPS-stimulated J774.1 mouse macrophages. Using an siRNA-mediated knockdown and specific antagonist, it was found that the ATP-induced enhanced inflammatory responses were mediated through P2X7R. We then screened 42 polyphenols for inhibiting the ATP/P2X7R-induced calcium influx, and found that several polyphenols exhibited significant inhibitory effects. Especially, a flavonoid baicalein significantly inhibited ATP-induced inflammation, including interleukin-1β secretion, through inhibition of the ATP/P2X7R signaling. These findings suggest that ATP/P2X7R signaling plays an important role in excess inflammatory responses and could be a potential anti-inflammatory target of natural polyphenolic compounds.
Collapse
|
162
|
Ott BR, Jones RN, Daiello LA, de la Monte SM, Stopa EG, Johanson CE, Denby C, Grammas P. Blood-Cerebrospinal Fluid Barrier Gradients in Mild Cognitive Impairment and Alzheimer's Disease: Relationship to Inflammatory Cytokines and Chemokines. Front Aging Neurosci 2018; 10:245. [PMID: 30186149 PMCID: PMC6110816 DOI: 10.3389/fnagi.2018.00245] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 07/25/2018] [Indexed: 01/30/2023] Open
Abstract
Background: The pathophysiology underlying altered blood-cerebrospinal fluid barrier (BCSFB) function in Alzheimer's disease (AD) is unknown but may relate to endothelial cell activation and cytokine mediated inflammation. Methods: Cerebrospinal fluid (CSF) and peripheral blood were concurrently collected from cognitively healthy controls (N = 21) and patients with mild cognitive impairment (MCI) (N = 8) or AD (N = 11). The paired serum and CSF samples were assayed for a panel of cytokines, chemokines, and related trophic factors using multiplex ELISAs. Dominance analysis models were conducted to determine the relative importance of the inflammatory factors in relationship to BCSFB permeability, as measured by CSF/serum ratios for urea, creatinine, and albumin. Results: BCSFB disruption to urea, a small molecule distributed by passive diffusion, had a full model coefficient of determination (r2) = 0.35, and large standardized dominance weights (>0.1) for monocyte chemoattractant protein-1, interleukin (IL)-15, IL-1rα, and IL-2 in serum. BCSFB disruption to creatinine, a larger molecule governed by active transport, had a full model r2 = 0.78, and large standardized dominance weights for monocyte inhibitor protein-1b in CSF and tumor necrosis factor-α in serum. BCSFB disruption to albumin, a much larger molecule, had a full model r2 = 0.62, and large standardized dominance weights for IL-17a, interferon-gamma, IL-2, and VEGF in CSF, as well IL-4 in serum. Conclusions: Inflammatory proteins have been widely documented in the AD brain. The results of the current study suggest that changes in BCSFB function resulting in altered permeability and transport are related to expression of specific inflammatory proteins, and that the shifting distribution of these proteins from serum to CSF in AD and MCI is correlated with more severe perturbations in BCSFB function.
Collapse
Affiliation(s)
- Brian R. Ott
- Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States,George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States,*Correspondence: Brian R. Ott
| | - Richard N. Jones
- Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Lori A. Daiello
- Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Suzanne M. de la Monte
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States,Division of Neuropathology, Department of Pathology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Edward G. Stopa
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States,Division of Neuropathology, Department of Pathology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Conrad E. Johanson
- Department of Neurosurgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Charles Denby
- Department of Neurology, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Paula Grammas
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States
| |
Collapse
|
163
|
Vérité J, Page G, Paccalin M, Julian A, Janet T. Differential chemokine expression under the control of peripheral blood mononuclear cells issued from Alzheimer's patients in a human blood brain barrier model. PLoS One 2018; 13:e0201232. [PMID: 30092003 PMCID: PMC6084889 DOI: 10.1371/journal.pone.0201232] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/11/2018] [Indexed: 12/26/2022] Open
Abstract
Growing evidence highlights the peripheral blood mononuclear cells (PBMCs) role and the chemokine involvement in the Alzheimer's disease (AD) physiopathology. However, few data are available about the impact of AD PBMCs in the chemokine signature in a brain with AD phenotype. Therefore, this study analyzed the chemokine levels in a human blood brain barrier model. A human endothelial cell line from the immortalized cerebral microvascular endothelial cell line (hCMEC/D3) and a human glioblastoma U-87 MG cell line, both with no AD phenotype were used while PBMCs came from AD at mild or moderate stage and control patients. PBMCs from moderate AD patients decreased CCL2 and CCL5 levels in endothelial, and also CXCL10 in abluminal compartments and in PBMCs compared to PBMCs from mild AD patients. The CX3CL1 expression increased in endothelial and abluminal compartments with PBMCs from mild AD patients compared to controls. AD PBMCs can convert the chemokine signature towards that found in AD brain, targeting some chemokines as new biomarkers in AD.
Collapse
Affiliation(s)
- Julie Vérité
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, Poitiers, France
| | - Guylène Page
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, Poitiers, France
| | - Marc Paccalin
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, Poitiers, France
- Department of Geriatrics, Poitiers University Hospital, Poitiers, France
- Memory Resource and Research Center of Poitiers, Poitiers University Hospital, Poitiers, France
| | - Adrien Julian
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, Poitiers, France
- Memory Resource and Research Center of Poitiers, Poitiers University Hospital, Poitiers, France
- Department of Neurology, Poitiers University Hospital, Poitiers, France
| | - Thierry Janet
- EA3808, molecular Targets and Therapeutics of Alzheimer’s disease, University of Poitiers, Poitiers, France
| |
Collapse
|
164
|
Xu X, Zhang A, Zhu Y, He W, Di W, Fang Y, Shi X. MFG-E8 reverses microglial-induced neurotoxic astrocyte (A1) via NF-κB and PI3K-Akt pathways. J Cell Physiol 2018; 234:904-914. [PMID: 30076715 DOI: 10.1002/jcp.26918] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 06/13/2018] [Indexed: 12/28/2022]
Abstract
Recent evidence have suggested that neuroinflammation and ischemia induce the activation of two different types of reactive astrocytes, termed A1 and A2. Additionally, A1 astrocytes contribute to the death of neurons and oligodendrocytes in neurodegenerative diseases, such as Alzheimer's disease (AD). In the current study, we constructed an Aβ42-activated microglia-conditioned medium to induce A1 astrocytic activation via secretion of interleukin 1α, tumor necrosis factor, and complement component 1q in vitro, and indicated the regulatory role of milk fat globule epidermal growth factor 8 (MFG-E8) on A1/A2 astrocytic alteration through the downregulation of nuclear factor-κB and the upregulation of PI3K-Akt. This study showed that MFG-E8 suppressed A1 astrocytes and holds great potential for the treatment of AD.
Collapse
Affiliation(s)
- Xiaotian Xu
- Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Aiwu Zhang
- Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingting Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wen He
- Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Di
- Department of Neurology, Shanxi Provincial People's Hospital, Xi'an, China.,Department of Neurology, Third Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Yannan Fang
- Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaolei Shi
- Department of Neurology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui Province, People's Republic of China.,Kinsmen Laboratory of Neurological Research, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
165
|
Montoliu-Gaya L, Güell-Bosch J, Esquerda-Canals G, Roda AR, Serra-Mir G, Lope-Piedrafita S, Sánchez-Quesada JL, Villegas S. Differential effects of apoE and apoJ mimetic peptides on the action of an anti-Aβ scFv in 3xTg-AD mice. Biochem Pharmacol 2018; 155:380-392. [PMID: 30026023 DOI: 10.1016/j.bcp.2018.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/13/2018] [Indexed: 12/31/2022]
Abstract
Anti-Aβ immunotherapy has emerged as a promising approach to treat Alzheimer's disease (AD). The single-chain variable fragment scFv-h3D6 is an anti-Aβ antibody fragment that lacks the Fc region, which is associated with the induction of microglial reactivity by the full-length monoclonal antibody bapineuzumab. ScFv-h3D6 was previously shown to restore the levels of apolipoprotein E (apoE) and apolipoprotein J (apoJ) in a triple-transgenic-AD (3xTg-AD) mouse model. Since apoE and apoJ play an important role in the development of AD, we aimed to study the in vivo effect of the combined therapy of scFv-h3D6 with apoE and apoJ mimetic peptides (MPs). Four-and-a-half-month-old 3xTg-AD mice were treated for six weeks with scFv-h3D6, apoE-MP, apoJ-MP, or a combination of scFv-h3D6 with each of the MPs, or a vehicle, and then the results were compared to non-transgenic mice. Magnetic Resonance Imaging showed a general tendency of the different treatments to protect against the reduction in brain volume. Aβ burden decreased after treatment with scFv-h3D6, apoE-MP, or apoJ-MP, but the effect was not as evident with the combined therapies. In terms of glial reactivity, apoE-MP showed a potent anti-inflammatory effect that was eased by the presence of scFv-h3D6, whereas the combination of apoJ-MP and scFv-h3D6 was not detrimental. ScFv-h3D6 alone did not induce microglial reactivity, as full-length antibodies do; rather, it reduced it. Endogenous apoE and apoJ levels were decreased by scFv-h3D6, but the MPs lead to a simultaneous increase of both apolipoproteins. While apoE-MP and apoJ-MP demonstrated different effects in the combined therapies with scFv-h3D6, they did not improve the overall protective effect of scFv-h3D6 in reducing the Aβ burden, apolipoproteins levels or microglial reactivity.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Jofre Güell-Bosch
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Gisela Esquerda-Canals
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Alejandro R Roda
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Gabriel Serra-Mir
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Silvia Lope-Piedrafita
- Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain; CIBER of Diabetes and Metabolic Diseases (CIBERDEM), Spain.
| | - Sandra Villegas
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| |
Collapse
|
166
|
Pogue AI, Lukiw WJ. Up-regulated Pro-inflammatory MicroRNAs (miRNAs) in Alzheimer's disease (AD) and Age-Related Macular Degeneration (AMD). Cell Mol Neurobiol 2018; 38:1021-1031. [PMID: 29302837 PMCID: PMC11481951 DOI: 10.1007/s10571-017-0572-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 12/21/2017] [Indexed: 11/25/2022]
Abstract
Alzheimer's disease (AD) of the brain neocortex and age-related macular degeneration (AMD) of the retina are two complex neurodegenerative disorders, which (i) involve the progressive dysregulation and deterioration of multiple neurobiological signaling pathways, (ii) exhibit the temporal accumulation of pro-inflammatory lesions including the amyloid beta (Aβ) peptide-containing senile plaques of AD and the drusen of AMD, and (iii) culminate in an insidious inflammatory neurodegeneration ending, respectively, in neural cell atrophy and death and progressive loss of cognition and central visual function. Recent independent research studies have indicated that AD and AMD share common, pathological signaling defects and disease mechanisms at the molecular genetic level. Using high-integrity total RNA samples pooled from AD brain and AMD retina, microfluidic hybridization miRNA arrays, and bioinformatics, the current study was undertaken to quantify microRNA (miRNA) speciation and complexity common to both AD and AMD. These small non-coding (sncRNAs) are known to post-transcriptionally regulate multiple neurobiological pathways and an abundance of research information has already been generated on the roles of these miRNAs in pathological situations involving inflammatory neuropathology and neural cell decline. Here, for the first time, we report the sequence and abundance of a septet of sncRNAs including miRNA-7, miRNA-9-1, miRNA-23a/miRNA-27a, miRNA-34a, miRNA-125b-1, miRNA-146a, and miRNA-155 that are significantly increased in abundance and common to both AD-affected superior temporal lobe neocortex (Brodmann A22) and the AMD-affected macular region of the retina. Bioinformatics, miRNA-mRNA complementarity, next-gen RNA sequencing, and feature alignment analysis further indicate that these 7 up-regulated miRNAs have the potential to interact with and down-regulate ~ 9460 target messenger RNAs (mRNAs; about 3.5% of the genome) involved in the synchronization of amyloid production and clearance, phagocytosis, innate-immune, pro-inflammatory, and neurotrophic signaling and/or synaptogenesis in diseased tissues.
Collapse
Affiliation(s)
| | - Walter J Lukiw
- Alchem Biotech Research, Toronto, ON, Canada.
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- Department of Neurology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- Department of Ophthalmology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112-2272, USA.
| |
Collapse
|
167
|
Khalili M, Alavi M, Esmaeil-Jamaat E, Baluchnejadmojarad T, Roghani M. Trigonelline mitigates lipopolysaccharide-induced learning and memory impairment in the rat due to its anti-oxidative and anti-inflammatory effect. Int Immunopharmacol 2018; 61:355-362. [PMID: 29935483 DOI: 10.1016/j.intimp.2018.06.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 06/05/2018] [Accepted: 06/12/2018] [Indexed: 12/15/2022]
Abstract
Brain inflammation is associated with cognitive dysfunction, especially in elderly. Trigonelline is a plant alkaloid and a major component of coffee and fenugreek with anti-diabetic, antioxidant, anti-inflammatory, and neuroprotective effects. In this study, the beneficial effect of trigonelline against lipopolysaccharide (LPS)-induced cognitive decline was assessed in the rat. LPS was injected i.p. at a dose of 500 μg/kg to induce neuroinflammation and trigonelline was administered p.o. at doses of 20, 40, or 80 mg/kg/day 1 h after LPS that continued for one week. Trigonelline-treated LPS-challenged rats showed improved spatial recognition memory in Y maze, discrimination ratio in novel object discrimination test, and retention and recall in passive avoidance paradigm. Additionally, trigonelline lowered hippocampal malondialdehyde (MDA) and acetylcholinesterase (AChE) activity and improved superoxide dismutase (SOD), catalase, and glutathione (GSH). Furthermore, trigonelline depressed hippocampal nuclear factor-kappaB (NF-κB), toll-like receptor 4 (TLR4), and tumor necrosis factor α (TNF α) in LPS-challenged rats. All of the effects of trigonelline followed a dose-dependent pattern and in some aspects, it acted even better than the routinely-used anti-inflammatory drug dexamethasone. Collectively, trigonelline is capable to diminish LPS-induced cognitive decline via suppression of hippocampal oxidative stress and inflammation and appropriate modulation of NF-κB/TLR4 and AChE activity.
Collapse
Affiliation(s)
- Mohsen Khalili
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| | - Mitra Alavi
- School of Medicine, Shahed University, Tehran, Iran
| | | | | | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| |
Collapse
|
168
|
Lanza G, Centonze SS, Destro G, Vella V, Bellomo M, Pennisi M, Bella R, Ciavardelli D. Shiatsu as an adjuvant therapy for depression in patients with Alzheimer's disease: A pilot study. Complement Ther Med 2018; 38:74-78. [PMID: 29857884 DOI: 10.1016/j.ctim.2018.04.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/24/2018] [Accepted: 04/30/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Among the complementary and alternative medicine, Shiatsu might represent a feasible option for depression in Alzheimer's disease (AD). We evaluated Shiatsu on mood, cognition, and functional independence in patients undergoing physical activity. DESIGN Single-blind randomized controlled study. SETTING Dedicated Community Center for patients with AD. INTERVENTIONS AD patients with depression were randomly assigned to the "active group" (Shiatsu + physical activity) or the "control group" (physical activity alone). Shiatsu was performed by the same therapist once a week for ten months. MAIN OUTCOME MEASURES Global cognitive functioning (Mini Mental State Examination - MMSE), depressive symptoms (Geriatric Depression Scale - GDS), and functional status (Activity of Daily Living - ADL, Instrumental ADL - IADL) were assessed before and after the intervention. RESULTS We found a within-group improvement of MMSE, ADL, and GDS in the active group. However, the analysis of differences before and after the interventions showed a statistically significant decrease of GDS score only in the active group. CONCLUSIONS The combination of Shiatsu and physical activity improved depression in AD patients compared to physical activity alone. The pathomechanism might involve neuroendocrine-mediated effects of Shiatsu on neural circuits implicated in mood and affect regulation.
Collapse
Affiliation(s)
| | | | - Gera Destro
- Centro diurno Alzheimer, Ospedale "Michele Chiello", ASP 4, Piazza Armerina, Italy
| | - Veronica Vella
- School of Human and Social Science, University "Kore" of Enna, Enna, Italy; Department of Clinical and Molecular Bio-Medicine, Endocrinology Unit, University of Catania "Garibaldi-Nesima Medical Center", Catania, Italy
| | - Maria Bellomo
- School of Human and Social Science, University "Kore" of Enna, Enna, Italy
| | - Manuela Pennisi
- Spinal Unit, Emergency Hospital "Cannizzaro", Catania, Italy
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies, Section of Neurosciences, Azienda Ospedaliero Universitaria Policlinico Vittorio Emanuele, Catania, Italy
| | - Domenico Ciavardelli
- School of Human and Social Science, University "Kore" of Enna, Enna, Italy; Centro Scienze dell'Invecchiamento e Medicina Traslazionale - CeSI-Met, Chieti, Italy
| |
Collapse
|
169
|
Dong S, Maniar S, Manole MD, Sun D. Cerebral Hypoperfusion and Other Shared Brain Pathologies in Ischemic Stroke and Alzheimer's Disease. Transl Stroke Res 2018; 9:238-250. [PMID: 28971348 PMCID: PMC9732865 DOI: 10.1007/s12975-017-0570-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/05/2017] [Accepted: 09/13/2017] [Indexed: 12/13/2022]
Abstract
Newly emerged evidence reveals that ischemic stroke and Alzheimer's disease (AD) share pathophysiological changes in brain tissue including hypoperfusion, oxidative stress, immune exhaustion, and inflammation. A mechanistic link between hypoperfusion and amyloid β accumulation can lead to cell damage as well as to motor and cognitive deficits. This review will discuss decreased cerebral perfusion and other related pathophysiological changes common to both ischemic stroke and AD, such as vascular damages, cerebral blood flow alteration, abnormal expression of amyloid β and tau proteins, as well as behavioral and cognitive deficits. Furthermore, this review highlights current treatment options and potential therapeutic targets that warrant further investigation.
Collapse
Affiliation(s)
- Shuying Dong
- Department of Pharmacology, Bengbu Medical College, Bengbu, Anhui, China
- Department of Neurology, University of Pittsburgh, S-598 South Biomedical Science Tower, 3500 Terrace St., Pittsburgh, PA, 15213, USA
| | - Shelly Maniar
- Department of Neurology, University of Pittsburgh, S-598 South Biomedical Science Tower, 3500 Terrace St., Pittsburgh, PA, 15213, USA
- Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, Pennsylvania, 15601, USA
| | - Mioara D Manole
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, S-598 South Biomedical Science Tower, 3500 Terrace St., Pittsburgh, PA, 15213, USA.
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, PA, USA.
| |
Collapse
|
170
|
Abstract
PURPOSE OF REVIEW Despite the extensive research carried out in the past decades, the current pathophysiological notions of neurodegenerative disease as well as effective treatments to reduce their progression are largely unknown. Alterations of the human microbiota, the plethora of different microscopic organisms that our body hosts, have been linked to neurodegenerative disease risk, onset and progression. This review summarizes the current knowledge on the possible role of microbiota in neurodegenerative disorders and briefly discusses strategies to restore microbiota homeostasis. RECENT FINDINGS Preclinical evidences and human cross-sectional studies posit the gut microbiota as a key actor in the Parkinson's disease onset and progression, reporting the presence of a specific gut microbiota profile in association with the modulation of disease and symptoms. Gut microbiota alterations have been correlated with brain disease and peripheral inflammation also in Alzheimer's patients. SUMMARY The interaction between the microbiota and the host is promising to answer clinical questions that have so far escaped clarification with the current pathophysiological notions of health and disease. However, human longitudinal studies starting in the earlier disease phases are needed to understand the causative relation between microbiota and the hallmarks of these neurodegenerative disorders and to develop innovative treatments aimed at preventing or slowing brain damages.
Collapse
|
171
|
Zhao Y, Lukiw WJ. Bacteroidetes Neurotoxins and Inflammatory Neurodegeneration. Mol Neurobiol 2018; 55:9100-9107. [PMID: 29637444 DOI: 10.1007/s12035-018-1015-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/16/2018] [Indexed: 12/31/2022]
Abstract
The gram-negative facultative anaerobe Bacteroides fragilis (B. fragilis) constitutes an appreciable proportion of the human gastrointestinal (GI)-tract microbiome. As is typical of most gram-negative bacilli, B. fragilis secretes an unusually complex mixture of neurotoxins including the extremely pro-inflammatory lipopolysaccharide BF-LPS. LPS (i) has recently been shown to associate with the periphery of neuronal nuclei in sporadic Alzheimer's disease (AD) brain and (ii) promotes the generation of the inflammatory transcription factor NF-kB (p50/p65 complex) in human neuronal-glial cells in primary-culture. In turn, the NF-kB (p50/p65 complex) strongly induces the transcription of a small family of pro-inflammatory microRNAs (miRNAs) including miRNA-9, miRNA-34a, miRNA-125b, miRNA-146a, and miRNA-155. These ultimately bind with the 3'-untranslated region (3'-UTR) of several target messenger RNAs (mRNAs) and thereby reduce their expression. Down-regulated mRNAs include those encoding complement factor-H (CFH), an SH3-proline-rich multi-domain-scaffolding protein of the postsynaptic density (SHANK3), and the triggering receptor expressed in myeloid/microglial cells (TREM2), as is observed in sporadic AD brain. Hence, a LPS normally confined to the GI tract is capable of driving a NF-kB-miRNA-mediated deficiency in gene expression that contributes to alterations in synaptic-architecture and synaptic-deficits, amyloidogenesis, innate-immune defects, and progressive inflammatory signaling, all of which are characteristics of AD-type neurodegeneration. This article will review the most recent research which supports the idea that bacterial components of the GI tract microbiome such as BF-LPS can transverse biophysical barriers and contribute to AD-type change. For the first-time, these results indicate that specific GI tract microbiome-derived neurotoxins have a strong pathogenic role in eliciting alterations in NF-kB-miRNA-directed gene expression that drives the AD process.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112, USA.,Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112, USA
| | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112, USA. .,Department of Neurology, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112, USA. .,Departments of Ophthalmology, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112, USA.
| |
Collapse
|
172
|
Julian A, Rioux-Bilan A, Ragot S, Krolak-Salmon P, Berrut G, Dantoine T, Hommet C, Hanon O, Page G, Paccalin M. Blood Inflammatory Mediators and Cognitive Decline in Alzheimer's Disease: A Two Years Longitudinal Study. J Alzheimers Dis 2018; 63:87-92. [PMID: 29614665 DOI: 10.3233/jad-171131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Peripheral inflammatory processes are involved in Alzheimer's disease (AD). We aimed to determine whether plasma inflammatory mediator levels at diagnosis are associated with cognitive decline through a 2-year follow-up in AD patients. Patients (n = 109, mean age 79.44 (6.82) years) were included at diagnosis with MMSE scores between 16 and 25, with C-reactive protein <10 mg/L, and without any acute or chronic inflammation status. Plasma IL-1β, IL-6, TNF-α, and CCL5 were measured using Luminex X-MAP technology at baseline, and after one year and two years of follow-up. The mean values of IL-1β, IL-6, TNF-α, and CCL5 at diagnosis were 0.3, 1.94, 6.57, and 69,615.81 pg/mL, respectively. Mean cognitive decline in MMSE was 3.35 points. No correlation between plasmatic value of IL-1β, IL-6, TNF-α, or CCL5 at diagnosis and cognitive decline during the two years of follow-up was found. Cognitive decline in AD does not appear to be predictable by the tested inflammatory mediators.
Collapse
Affiliation(s)
- Adrien Julian
- Poitiers University Hospital, Centre Mémoire de Ressources et de Recherche, France.,Department of Neurology, Poitiers University Hospital, France.,University of Poitiers, EA3808 Molecular Targets and Therapeutic of Alzheimer's disease, Poitiers, France
| | - Agnès Rioux-Bilan
- University of Poitiers, EA3808 Molecular Targets and Therapeutic of Alzheimer's disease, Poitiers, France
| | - Stéphanie Ragot
- Poitiers University Hospital, Centre d'Investigation Clinique, France
| | | | - Gilles Berrut
- Department of Geriatrics, Nantes University Hospital, France
| | | | - Caroline Hommet
- Tours University Hospital, Centre Mémoire de Ressources et de Recherche, France
| | - Olivier Hanon
- Department of Geriatrics, Paris Broca University Hospital, France
| | - Guylène Page
- University of Poitiers, EA3808 Molecular Targets and Therapeutic of Alzheimer's disease, Poitiers, France
| | - Marc Paccalin
- Poitiers University Hospital, Centre Mémoire de Ressources et de Recherche, France.,University of Poitiers, EA3808 Molecular Targets and Therapeutic of Alzheimer's disease, Poitiers, France.,Poitiers University Hospital, Centre d'Investigation Clinique, France.,Department of Geriatrics, Poitiers University Hospital, France
| |
Collapse
|
173
|
Mirahmadi SMS, Shahmohammadi A, Rousta AM, Azadi MR, Fahanik-Babaei J, Baluchnejadmojarad T, Roghani M. Soy isoflavone genistein attenuates lipopolysaccharide-induced cognitive impairments in the rat via exerting anti-oxidative and anti-inflammatory effects. Cytokine 2018; 104:151-159. [PMID: 29102164 DOI: 10.1016/j.cyto.2017.10.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/04/2017] [Accepted: 10/08/2017] [Indexed: 12/16/2022]
Abstract
Systemic inflammation during infectious disorders usually accompanies chronic complications including cognitive dysfunction. Neuroinflammation and cognitive deficit are also observed in some debilitating neurological disorders like Alzheimer's and Parkinson's diseases. Genistein is a soy isoflavone with multiple beneficial effects including anti-inflammatory, anti-oxidative, and protective properties. In this research study, the effect of genistein in prevention of lipopolysaccharide (LPS)-induced cognitive dysfunction was investigated. LPS was given i.p. (500 μg/kg/day) and genistein was orally given (10, 50, or 100 mg/kg) for one week. Findings showed that genistein could dose-dependently attenuate spatial recognition, discrimination, and memory deficits. Additionally, genistein treatment of LPS-challenged group lowered hippocampal level of malondialdehyde (MDA) and increased activity of superoxide dismutase (SOD) and catalase and glutathione (GSH) level. Furthermore, genistein ameliorated hippocampal acetylcholinesterase (AChE) activity in LPS-challenged rats. Furthermore, genistein administration to LPS-injected group lowered hippocampal level of interleukin 6 (IL-6), nuclear factor-kappaB (NF-κB) p65, toll-like receptor 4 (TLR4), tumor necrosis factor α (TNFα), cyclooxygenase-2 (COX2), inducible nitric oxide synthase (iNOS), glial fibrillary acidic protein (GFAP), and increased hippocampal level of antioxidant element nuclear factor (erythroid-derived 2)-like 2 (Nrf2). In conclusion, genistein alleviated LPS-induced cognitive dysfunctions and neural inflammation attenuation of oxidative stress and AChE activity and appropriate modulation of Nrf2/NF-κB/IL-6/TNFα/COX2/iNOS/TLR4/GFAP.
Collapse
Affiliation(s)
| | | | | | | | - Javad Fahanik-Babaei
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| |
Collapse
|
174
|
A Novel Association of Polymorphism in the ITGA4 Gene Encoding the VLA-4 α4 Subunit with Increased Risk of Alzheimer's Disease. Mediators Inflamm 2018; 2018:7623823. [PMID: 29769839 PMCID: PMC5892238 DOI: 10.1155/2018/7623823] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/11/2018] [Accepted: 02/28/2018] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia in elderly people worldwide. Many studies support the hypothesis that the inflammation of the CNS contributes to the neurodegeneration and disease progression. The integrin molecule α4β1, also known as very late antigen 4 (VLA-4), belongs to adhesion molecules that activate the inflammatory process through the migration of immune cells into the CNS. Therefore, the objective of our study was to analyze the association between two polymorphisms located in the ITGA4 gene encoding the α4 subunit of VLA-4 and the risk of AD. 104 late-onset AD patients and 206 control subjects from Slovakia were genotyped for ITGA4 gene SNP polymorphism rs113276800 (-269C/A) and rs1143676 (+3061A/G). The same study cohorts were also genotyped for the APOE-ε4, which is a known genetic factor associated with increased risk of AD developing. ITGA4 polymorphism analysis revealed significantly higher frequency of the +3061AG carriers in AD group compared to the controls (P ≤ 0.05). Following the APOE-ε4 stratification of study groups, the association remained significant only in APOE-ε4 noncarriers. Our study suggests a novel association of ITGA4 +3061A/G polymorphism with AD and its possible contribution to the disease pathology.
Collapse
|
175
|
Small GW, Siddarth P, Li Z, Miller KJ, Ercoli L, Emerson ND, Martinez J, Wong KP, Liu J, Merrill DA, Chen ST, Henning SM, Satyamurthy N, Huang SC, Heber D, Barrio JR. Memory and Brain Amyloid and Tau Effects of a Bioavailable Form of Curcumin in Non-Demented Adults: A Double-Blind, Placebo-Controlled 18-Month Trial. Am J Geriatr Psychiatry 2018; 26:266-277. [PMID: 29246725 DOI: 10.1016/j.jagp.2017.10.010] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/08/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Because curcumin's anti-inflammatory properties may protect the brain from neurodegeneration, we studied its effect on memory in non-demented adults and explored its impact on brain amyloid and tau accumulation using 2-(1-{6-[(2-[F-18]fluoroethyl)(methyl)amino]-2-naphthyl}ethylidene)malononitrile positron emission tomography (FDDNP-PET). METHODS Forty subjects (age 51-84 years) were randomized to a bioavailable form of curcumin (Theracurmin® containing 90 mg of curcumin twice daily [N = 21]) or placebo (N = 19) for 18 months. Primary outcomes were verbal (Buschke Selective Reminding Test [SRT]) and visual (Brief Visual Memory Test-Revised [BVMT-R]) memory, and attention (Trail Making A) was a secondary outcome. FDDNP-PET signals (15 curcumin, 15 placebo) were determined in amygdala, hypothalamus, medial and lateral temporal, posterior cingulate, parietal, frontal, and motor (reference) regions. Mixed effects general linear models controlling for age and education, and effect sizes (ES; Cohen's d) were estimated. RESULTS SRT Consistent Long-Term Retrieval improved with curcumin (ES = 0.63, p = 0.002) but not with placebo (ES = 0.06, p = 0.8; between-group: ES = 0.68, p = 0.05). Curcumin also improved SRT Total (ES = 0.53, p = 0.002), visual memory (BVMT-R Recall: ES = 0.50, p = 0.01; BVMT-R Delay: ES = 0.51, p = 0.006), and attention (ES = 0.96, p < 0.0001) compared with placebo (ES = 0.28, p = 0.1; between-group: ES = 0.67, p = 0.04). FDDNP binding decreased significantly in the amygdala with curcumin (ES = -0.41, p = 0.04) compared with placebo (ES = 0.08, p = 0.6; between-group: ES = 0.48, p = 0.07). In the hypothalamus, FDDNP binding did not change with curcumin (ES = -0.30, p = 0.2), but increased with placebo (ES = 0.26, p = 0.05; between-group: ES = 0.55, p = 0.02). CONCLUSIONS Daily oral Theracurmin may lead to improved memory and attention in non-demented adults. The FDDNP-PET findings suggest that symptom benefits are associated with decreases in amyloid and tau accumulation in brain regions modulating mood and memory.
Collapse
Affiliation(s)
- Gary W Small
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA.
| | - Prabha Siddarth
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Zhaoping Li
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Karen J Miller
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Linda Ercoli
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Natacha D Emerson
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Jacqueline Martinez
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Koon-Pong Wong
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Jie Liu
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - David A Merrill
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Stephen T Chen
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Susanne M Henning
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Nagichettiar Satyamurthy
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Sung-Cheng Huang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - David Heber
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| | - Jorge R Barrio
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, UCLA Longevity Center, Department of Molecular and Medical Pharmacology, and Center for Human Nutrition, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
176
|
Rahimi F. Aptamers Selected for Recognizing Amyloid β-Protein-A Case for Cautious Optimism. Int J Mol Sci 2018; 19:ijms19030668. [PMID: 29495486 PMCID: PMC5877529 DOI: 10.3390/ijms19030668] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/18/2018] [Accepted: 02/22/2018] [Indexed: 02/07/2023] Open
Abstract
Aptamers are versatile oligonucleotide ligands used for molecular recognition of diverse targets. However, application of aptamers to the field of amyloid β-protein (Aβ) has been limited so far. Aβ is an intrinsically disordered protein that exists in a dynamic conformational equilibrium, presenting time-dependent ensembles of short-lived, metastable structures and assemblies that have been generally difficult to isolate and characterize. Moreover, despite understanding of potential physiological roles of Aβ, this peptide has been linked to the pathogenesis of Alzheimer disease, and its pathogenic roles remain controversial. Accumulated scientific evidence thus far highlights undesirable or nonspecific interactions between selected aptamers and different Aβ assemblies likely due to the metastable nature of Aβ or inherent affinity of RNA oligonucleotides to β-sheet-rich fibrillar structures of amyloidogenic proteins. Accordingly, lessons drawn from Aβ–aptamer studies emphasize that purity and uniformity of the protein target and rigorous characterization of aptamers’ specificity are important for realizing and garnering the full potential of aptamers selected for recognizing Aβ or other intrinsically disordered proteins. This review summarizes studies of aptamers selected for recognizing different Aβ assemblies and highlights controversies, difficulties, and limitations of such studies.
Collapse
Affiliation(s)
- Farid Rahimi
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|
177
|
Li W, Chai Q, Zhang H, Ma J, Xu C, Dong J, Wei X, Wang Z, Zhang K. High doses of minocycline may induce delayed activation of microglia in aged rats and thus cannot prevent postoperative cognitive dysfunction. J Int Med Res 2018; 46:1404-1413. [PMID: 29458276 PMCID: PMC6091817 DOI: 10.1177/0300060517754032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Objective Postoperative cognitive dysfunction (POCD) is common after surgery in elderly patients and is associated with high morbidity. The molecular mechanisms responsible for POCD are unknown. Minocycline, an inhibitor of microglial activation, may be useful in treating and preventing POCD. We explored whether minocycline can inhibit microglial activation and prevent POCD in aged rats as a surgery model. Methods Rats aged 18 to 20 months were randomly allocated to the following groups: naïve, abdominal surgery alone, or minocycline injection before abdominal surgery. Hippocampal cytokine mRNA levels were measured at 3 hours, 1 day, 3 days, and 7 days after surgery, and microglial activation was measured at 3 hours and 7 days after surgery. Memory was assessed using the Morris water maze test. Results Surgery resulted in severe cognitive impairment in aged rats and induced a significant neuroinflammatory response and microglial activation. The use of minocycline can prevent microglial activation after surgery, but delayed microglial activation may occur. The use of minocycline may further impair memory after surgery. Conclusion Minocycline can restrain microglial activation and restrict the inflammatory response in the hippocampus early after surgery, but it may induce delayed microglial activation and cannot prevent POCD in aged rats.
Collapse
Affiliation(s)
- Wenyao Li
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qing Chai
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hongwei Zhang
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Ma
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chengfen Xu
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jifu Dong
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xianghua Wei
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhiyi Wang
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Kexian Zhang
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
178
|
Giau VV, Lee H, Shim KH, Bagyinszky E, An SSA. Genome-editing applications of CRISPR-Cas9 to promote in vitro studies of Alzheimer's disease. Clin Interv Aging 2018; 13:221-233. [PMID: 29445268 PMCID: PMC5808714 DOI: 10.2147/cia.s155145] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Genetic variations play an important role in the clinical presentation and progression of Alzheimer’s disease (AD), especially early-onset Alzheimer’s disease. Hundreds of mutations have been reported with the majority resulting from alterations in β-amyloid precursor protein (APP), presenilin 1 (PSEN1), or presenilin 2 (PSEN2) genes. The roles of these mutations in the pathogenesis of AD have been classically confirmed or refuted through functional studies, where the mutations are cloned, inserted into cell lines, and monitored for changes in various properties including cell survival, amyloid production, or Aβ42/40 ratio. However, these verification studies tend to be expensive, time consuming, and inconsistent. Recently, the clustered regularly interspaced short palindromic repeats-CRISPR-associated protein 9 (CRISPR–Cas9) system was developed, which improves sequence-specific gene editing in cell lines, organs, and animals. CRISPR–Cas9 is a promising tool for the generation of models of human genetic diseases and could facilitate the establishment of new animal AD models and the observation of dynamic bioprocesses in AD. Here, we recapitulated the history of CRISPR technology, recent progress, and, especially, its potential applications in AD-related genetic, animal modeling, and functional studies.
Collapse
Affiliation(s)
- Vo Van Giau
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Hyon Lee
- Department of Neurology, Gachon University Gil Medical Center, Incheon, South Korea
| | - Kyu Hwan Shim
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Eva Bagyinszky
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| |
Collapse
|
179
|
Zahr NM. Peripheral TNFα elevations in abstinent alcoholics are associated with hepatitis C infection. PLoS One 2018; 13:e0191586. [PMID: 29408932 PMCID: PMC5800541 DOI: 10.1371/journal.pone.0191586] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/08/2018] [Indexed: 12/31/2022] Open
Abstract
Substantial evidence supports the view that inflammatory processes contribute to brain alterations in HIV infection. Mechanisms recently proposed to underlie neuropathology in Alcohol Use Disorder (AUD) include elevations in peripheral cytokines that sensitize the brain to the damaging effects of alcohol. This study included 4 groups: healthy controls, individuals with AUD (abstinent from alcohol at examination), those infected with HIV, and those comorbid for HIV and AUD. The aim was to determine whether inflammatory cytokines are elevated in AUD as they are in HIV infection. Cytokines showing group differences included interferon gamma-induced protein 10 (IP-10) and tumor necrosis factor α (TNFα). Follow-up t-tests revealed that TNFα and IP-10 were higher in AUD than controls but only in AUD patients who were seropositive for Hepatitis C virus (HCV). Specificity of TNFα and IP-10 elevations to HCV infection status was provided by correlations between cytokine levels and HCV viral load and indices of liver integrity including albumin/globulin ratio, fibrosis scores, and AST/platelet count ratio. Because TNFα levels were mediated by HCV infection, this study provides no evidence for elevations in peripheral cytokines in "uncomplicated", abstinent alcoholics, independent of liver disease or HCV infection. Nonetheless, these results corroborate evidence for elevations in IP-10 and TNFα in HIV and for IP-10 levels in HIV+HCV co-infection.
Collapse
Affiliation(s)
- Natalie M. Zahr
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States of America
- Neuroscience Department, SRI International, Menlo Park, CA, United States of America
- * E-mail:
| |
Collapse
|
180
|
Abstract
The fundamental pathology in Alzheimer's disease (AD) is neuronal dysfunction leading to cognitive impairment. The amyloid-β peptide (Aβ), derived from amyloid precursor protein, is one driver of AD, but how it leads to neuronal dysfunction is not established. In this Review, I discuss the complexity of AD and possible cause-and-effect relationships between Aβ and the vascular and hemostatic systems. AD can be considered a multifactorial syndrome with various contributing pathological mechanisms. Therefore, as is routinely done with cancer, it will be important to classify patients with respect to their disease signature so that specific pathologies, including vascular pathways, can be therapeutically targeted.
Collapse
|
181
|
Wu SH, Lu IC, Lee SS, Kwan AL, Chai CY, Huang SH. Erythropoietin attenuates motor neuron programmed cell death in a burn animal model. PLoS One 2018; 13:e0190039. [PMID: 29385149 PMCID: PMC5791978 DOI: 10.1371/journal.pone.0190039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023] Open
Abstract
Burn-induced neuromuscular dysfunction may contribute to long-term morbidity; therefore, it is imperative to develop novel treatments. The present study investigated whether erythropoietin (EPO) administration attenuates burn-induced motor neuron apoptosis and neuroinflammatory response. To validate our hypothesis, a third-degree hind paw burn rat model was developed by bringing the paw into contact with a metal surface at 75°C for 10 s. A total of 24 male Sprague–Dawley rats were randomly assigned to four groups: Group A, sham-control; Group B, burn-induced; Group C, burn + single EPO dose (5000 IU/kg i.p. at D0); and Group D, burn + daily EPO dosage (3000 IU/kg/day i.p. at D0–D6). Two treatment regimens were used to evaluate single versus multiple doses treatment effects. Before sacrifice, blood samples were collected for hematological parameter examination. The histological analyses of microglia activation, iNOS, and COX-2 in the spinal cord ventral horn were performed at week 1 post-burn. In addition, we examined autophagy changes by biomarkers of LC3B and ATG5. The expression of BCL-2, BAX, cleaved caspase-3, phospho-AKT, and mTOR was assessed simultaneously through Western blotting. EPO administration after burn injury attenuated neuroinflammation through various mechanisms, including the reduction of microglia activity as well as iNOS and COX-2 expression in the spinal cord ventral horn. In addition, the expression of phospho-AKT, mTOR and apoptotic indicators, such as BAX, BCL-2, and cleaved caspase-3, was modulated. Furthermore, the activity of burn-induced autophagy in the spinal cord ventral horn characterized by the expression of autophagic biomarkers, LC3B and ATG5, was reduced after EPO administration. The present results indicate that EPO inhibits the AKT-mTOR pathway to attenuate burn-induced motor neuron programmed cell death and microglia activation. EPO can modulate neuroinflammation and programmed cell death and may be a therapeutic candidate for neuroprotection.
Collapse
Affiliation(s)
- Sheng-Hua Wu
- Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Anesthesiology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Cheng Lu
- Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Anesthesiology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Su-Shin Lee
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Hung Huang
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
182
|
Giulietti G, Torso M, Serra L, Spanò B, Marra C, Caltagirone C, Cercignani M, Bozzali M. Whole brain white matter histogram analysis of diffusion tensor imaging data detects microstructural damage in mild cognitive impairment and alzheimer's disease patients. J Magn Reson Imaging 2018; 48:767-779. [PMID: 29356183 DOI: 10.1002/jmri.25947] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 12/21/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Amnestic mild cognitive impairment (MCI) is a transitional stage between normal aging and Alzheimer's disease (AD). However, the clinical conversion from MCI to AD is unpredictable. Hence, identification of noninvasive biomarkers able to detect early changes induced by dementia is a pressing need. PURPOSE To explore the added value of histogram analysis applied to measures derived from diffusion tensor imaging (DTI) for detecting brain tissue differences between AD, MCI, and healthy subjects (HS). STUDY TYPE Prospective. POPULATION/SUBJECTS A local cohort (57 AD, 28 MCI, 23 HS), and an Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort (41 AD, 58 MCI, 41 HS). FIELD STRENGTH 3T. Dual-echo turbo spin echo (TSE); fluid-attenuated inversion recovery (FLAIR); modified-driven-equilibrium-Fourier-transform (MDEFT); inversion-recovery spoiled gradient recalled (IR-SPGR); diffusion tensor imaging (DTI). ASSESSMENT Normal-appearing white matter (NAWM) masks were obtained using the T1 -weighted volumes for tissue segmentation and T2 -weighted images for removal of hyperintensities/lesions. From DTI images, fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AXD), and radial diffusivity (RD) were obtained. NAWM histograms of FA, MD, AXD, and RD were derived and characterized estimating: peak height, peak location, mean value (MV), and quartiles (C25, C50, C75), which were compared between groups. Receiver operating characteristic (ROC) and area under ROC curves (AUC) were calculated. To confirm our results, the same analysis was repeated on the ADNI dataset. STATISTICAL TESTS One-way analysis of variance (ANOVA), post-hoc Student's t-test, multiclass ROC analysis. RESULTS For the local cohort, C25 of AXD had the maximum capability of group discrimination with AUC of 0.80 for "HS vs. patients" comparison and 0.74 for "AD vs. others" comparison. For the ADNI cohort, MV of AXD revealed the maximum group discrimination capability with AUC of 0.75 for "HS vs. patients" comparison and 0.75 for "AD vs. others" comparison. DATA CONCLUSION AXD of NAWM might be an early marker of microstructural brain tissue changes occurring during the AD course and might be useful for assessing disease progression. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2017.
Collapse
Affiliation(s)
| | - Mario Torso
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Laura Serra
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Barbara Spanò
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Camillo Marra
- Institute of Neurology, Catholic University, Rome, Italy
| | - Carlo Caltagirone
- Department of Systemic Medicine, University of Tor Vergata, Rome, Italy
- Clinical and Behavioral Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Mara Cercignani
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Neuroscience, Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, University of Sussex, Brighton, East Sussex, UK
| | - Marco Bozzali
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
183
|
Chen ZJ, Yang YF, Zhang YT, Yang DH. Dietary Total Prenylflavonoids from the Fruits of Psoralea corylifolia L. Prevents Age-Related Cognitive Deficits and Down-Regulates Alzheimer's Markers in SAMP8 Mice. Molecules 2018; 23:E196. [PMID: 29346315 PMCID: PMC6017019 DOI: 10.3390/molecules23010196] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is a serious threat for the aging society. In this study, we examined the preventive effect of the total prenylflavonoids (TPFB) prepared from the dried fruits of Psoralea corylifolia L., using an age-related AD mouse model SAMP8. We found that long-term dietary TPFB at 50 mg/kg·day significantly improved cognitive performance of the SAMP8 mice in Morris water maze tests, similar to 150 mg/kg·day of resveratrol, a popular neuro-protective compound. Furthermore, TPFB treatment showed significant improvements in various AD markers in SAMP8 brains, which were restored to near control levels of the normal mice, SAMR1. TPFB significantly reduced the level of amyloid β-peptide 42 (Aβ42), inhibited hyperphosphorylation of the microtubule-associated protein Tau, induced phosphorylation of Ser9 of the glycogen synthase kinase 3β (GSK-3β), and decreased the expression of the proinflammatory cytokines TNFα, IL-6, and IL-1β. Finally, TPFB also markedly reduced the level of serum derivatives of reactive oxygen metabolites (d-ROMs), a biomarker of oxidative stress in vivo. These results showed that dietary TPFB could effectively prevent age-related cognitive deficits and AD-like neurobiochemical changes, and may have a potential role in the prevention of Alzheimer's disease.
Collapse
Affiliation(s)
- Zhi-Jing Chen
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Yan-Fang Yang
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Ying-Tao Zhang
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Dong-Hui Yang
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
184
|
Gu Y, Ma LJ, Bai XX, Jie J, Zhang XF, Chen D, Li XP. Mitogen-activated protein kinase phosphatase 1 protects PC12 cells from amyloid beta-induced neurotoxicity. Neural Regen Res 2018; 13:1842-1850. [PMID: 30136701 PMCID: PMC6128043 DOI: 10.4103/1673-5374.238621] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The mitogen-activated protein kinase (MAPK) signaling pathway plays an important role in the regulation of cell growth, proliferation, differentiation, transformation and death. Mitogen-activated protein kinase phosphatase 1 (MKP1) has an inhibitory effect on the p38MAPK and JNK pathways, but it is unknown whether it plays a role in Aβ-induced oxidative stress and neuronal inflammation. In this study, PC12 cells were infected with MKP1 shRNA, MKP1 lentivirus or control lentivirus for 12 hours, and then treated with 0.1, 1, 10 or 100 μM amyloid beta 42 (Aβ42). The cell survival rate was measured using the cell counting kit-8 assay. MKP1, tumor necrosis factor-alpha (TNF-α) and interleukin-1β (IL-1β) mRNA expression levels were analyzed using quantitative real time-polymerase chain reaction. MKP1 and phospho-c-Jun N-terminal kinase (JNK) expression levels were assessed using western blot assay. Reactive oxygen species (ROS) levels were detected using 2′,7′-dichlorofluorescein diacetate. Mitochondrial membrane potential was measured using flow cytometry. Superoxide dismutase activity and malondialdehyde levels were evaluated using the colorimetric method. Lactate dehydrogenase activity was measured using a microplate reader. Caspase-3 expression levels were assessed by enzyme-linked immunosorbent assay. Apoptosis was evaluated using the terminal deoxynucleotidyl transferase dUTP nick end labeling method. MKP1 overexpression inhibited Aβ-induced JNK phosphorylation and the increase in ROS levels. It also suppressed the Aβ-induced increase in TNF-α and IL-1β levels as well as apoptosis in PC12 cells. In contrast, MKP1 knockdown by RNA interference aggravated Aβ-induced oxidative stress, inflammation and cell damage in PC12 cells. Furthermore, the JNK-specific inhibitor SP600125 abolished this effect of MKP1 knockdown on Aβ-induced neurotoxicity. Collectively, these results show that MKP1 mitigates Aβ-induced apoptosis, oxidative stress and neuroinflammation by inhibiting the JNK signaling pathway, thereby playing a neuroprotective role.
Collapse
Affiliation(s)
- Yue Gu
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Lian-Jun Ma
- Endoscopy Center, the China-Japan Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiao-Xue Bai
- Cadre's Wards, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jing Jie
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiu-Fang Zhang
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Dong Chen
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiao-Ping Li
- Department of Pediatrics, the First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
185
|
Zhao Y, Cong L, Lukiw WJ. Plant and Animal microRNAs (miRNAs) and Their Potential for Inter-kingdom Communication. Cell Mol Neurobiol 2018; 38:133-140. [PMID: 28879580 PMCID: PMC11482019 DOI: 10.1007/s10571-017-0547-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/31/2017] [Indexed: 12/22/2022]
Abstract
microRNAs (miRNAs) comprise a class of ~18-25 nucleotide (nt) single-stranded non-coding RNAs (sncRNAs) that are the smallest known carriers of gene-encoded, post-transcriptional regulatory information in both plants and animals. There are many fundamental similarities between plant and animal miRNAs-the miRNAs of both kingdoms play essential roles in development, aging and disease, and the shaping of the transcriptome of many cell types. Both plant and animal miRNAs appear to predominantly exert their genetic and transcriptomic influences by regulating gene expression at the level of messenger RNA (mRNA) stability and/or translational inhibition. Certain miRNA species, such as miRNA-155, miRNA-168, and members of the miRNA-854 family may be expressed in both plants and animals, suggesting a common origin and functional selection of specific miRNAs over vast periods of evolution (for example, Arabidopsis thaliana-Homo sapiens divergence ~1.5 billion years). Although there is emerging evidence for cross-kingdom miRNA communication-that plant-enriched miRNAs may enter the diet and play physiological and/or pathophysiological roles in human health and disease-some research reports repudiate this possibility. This research paper highlights some recent, controversial, and remarkable findings in plant- and animal-based miRNA signaling research with emphasis on the intriguing possibility that dietary miRNAs and/or sncRNAs may have potential to contribute to both intra- and inter-kingdom signaling, and in doing so modulate molecular-genetic mechanisms associated with human health and disease.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112-2272, USA
- Department of Anatomy and Cell Biology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA
| | - Lin Cong
- LSU Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112-2272, USA
- Department of Neurology, Shengjing Hospital, China Medical University, 36 No. 3 Street, Heping District, Shenyang, Liaoning, China
| | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112-2272, USA.
- Department of Neurology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- Department of Ophthalmology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
| |
Collapse
|
186
|
Terron A, Bal-Price A, Paini A, Monnet-Tschudi F, Bennekou SH, Leist M, Schildknecht S. An adverse outcome pathway for parkinsonian motor deficits associated with mitochondrial complex I inhibition. Arch Toxicol 2018; 92:41-82. [PMID: 29209747 PMCID: PMC5773657 DOI: 10.1007/s00204-017-2133-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022]
Abstract
Epidemiological studies have observed an association between pesticide exposure and the development of Parkinson's disease, but have not established causality. The concept of an adverse outcome pathway (AOP) has been developed as a framework for the organization of available information linking the modulation of a molecular target [molecular initiating event (MIE)], via a sequence of essential biological key events (KEs), with an adverse outcome (AO). Here, we present an AOP covering the toxicological pathways that link the binding of an inhibitor to mitochondrial complex I (i.e., the MIE) with the onset of parkinsonian motor deficits (i.e., the AO). This AOP was developed according to the Organisation for Economic Co-operation and Development guidelines and uploaded to the AOP database. The KEs linking complex I inhibition to parkinsonian motor deficits are mitochondrial dysfunction, impaired proteostasis, neuroinflammation, and the degeneration of dopaminergic neurons of the substantia nigra. These KEs, by convention, were linearly organized. However, there was also evidence of additional feed-forward connections and shortcuts between the KEs, possibly depending on the intensity of the insult and the model system applied. The present AOP demonstrates mechanistic plausibility for epidemiological observations on a relationship between pesticide exposure and an elevated risk for Parkinson's disease development.
Collapse
Affiliation(s)
| | | | - Alicia Paini
- European Commission Joint Research Centre, Ispra, Italy
| | | | | | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstr. 10, PO Box M657, 78457, Konstanz, Germany
| | - Stefan Schildknecht
- In Vitro Toxicology and Biomedicine, Department of Biology, University of Konstanz, Universitätsstr. 10, PO Box M657, 78457, Konstanz, Germany.
| |
Collapse
|
187
|
Role of the peripheral innate immune system in the development of Alzheimer's disease. Exp Gerontol 2017; 107:59-66. [PMID: 29275160 DOI: 10.1016/j.exger.2017.12.019] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/27/2017] [Accepted: 12/20/2017] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease is one of the most devastating neurodegenerative diseases. The exact cause of the disease is still not known although many scientists believe in the beta amyloid hypothesis which states that the accumulation of the amyloid peptide beta (Aβ) in brain is the initial cause which consequently leads to pathological neuroinflammation. However, it was recently shown that Aβ may have an important role in defending the brain against infections. Thus, the balance between positive and negative impact of Aβ may determine disease progression. Microglia in the brain are innate immune cells, and brain-initiated inflammatory responses reflected in the periphery suggests that Alzheimer's disease is to some extent also a systemic inflammatory disease. Greater permeability of the blood brain barrier facilitates the transport of peripheral immune cells to the brain and vice versa so that a vicious circle originating on the periphery may contribute to the development of overt clinical AD. Persistent inflammatory challenges by pathogens in the periphery, increasing with age, may also contribute to the central propagation of the pathological changes seen clinically. Therefore, the activation status of peripheral innate immune cells may represent an early biomarker of the upcoming impact on the brain. The modulation of these cells may thus become a useful mechanism for modifying disease progression.
Collapse
|
188
|
Zhao Y, Cong L, Lukiw WJ. Lipopolysaccharide (LPS) Accumulates in Neocortical Neurons of Alzheimer's Disease (AD) Brain and Impairs Transcription in Human Neuronal-Glial Primary Co-cultures. Front Aging Neurosci 2017; 9:407. [PMID: 29311897 PMCID: PMC5732913 DOI: 10.3389/fnagi.2017.00407] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/24/2017] [Indexed: 11/26/2022] Open
Abstract
Several independent laboratories have recently reported the detection of bacterial nucleic acid sequences or bacterial-derived neurotoxins, such as highly inflammatory lipopolysaccharide (LPS), within Alzheimer’s disease (AD) affected brain tissues. Whether these bacterial neurotoxins originate from the gastrointestinal (GI) tract microbiome, a possible brain microbiome or some dormant pathological microbiome is currently not well understood. Previous studies indicate that the co-localization of pro-inflammatory LPS with AD-affected brain cell nuclei suggests that there may be a contribution of this neurotoxin to genotoxic events that support inflammatory neurodegeneration and failure in homeostatic gene expression. In this report we provide evidence that in sporadic AD, LPS progressively accumulates in neuronal parenchyma and appears to preferentially associate with the periphery of neuronal nuclei. Run-on transcription studies utilizing [α-32P]-uridine triphosphate incorporation into newly synthesized total RNA further indicates that human neuronal-glial (HNG) cells in primary co-culture incubated with LPS exhibit significantly reduced output of DNA transcription products. These studies suggest that in AD LPS may impair the efficient readout of neuronal genetic information normally required for the homeostatic operation of brain cell function and may contribute to a progressive disruption in the read-out of genetic information.
Collapse
Affiliation(s)
- Yuhai Zhao
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Departments of Anatomy and Cell Biology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Lin Cong
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Neurology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Walter J Lukiw
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Neurology, Shengjing Hospital, China Medical University, Shenyang, China.,Department of Neurology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Ophthalmology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
189
|
Can small molecule inhibitors of glutaminyl cyclase be used as a therapeutic for Alzheimer's disease? Future Med Chem 2017; 9:1979-1981. [DOI: 10.4155/fmc-2017-0190] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial and socioeconomically burdensome disease. In view of the failures of anti-AD candidates, we should try to rethink what we did before and what we should do next, in part at least. Research shows that the more neurotoxic factor, pyroglutamate-Aβs, and the more important inflammatory mediators, pyroglutamate-CCL2, both contribute to the initiation of AD specifically and the generation of N-terminal intramolecular cyclization catalyzed by glutaminyl cyclase quality control, the over-expression of which correlates positively with the severity of AD. Subsequently, lowering pyroglutamate-Aβs and pyroglutamate-CCL2 levels by quality control inhibition using small molecule inhibitors could be expected as an amazing strategy for the prevention and treatment of AD.
Collapse
|
190
|
Modulation of the Senescence-Associated Inflammatory Phenotype in Human Fibroblasts by Olive Phenols. Int J Mol Sci 2017; 18:ijms18112275. [PMID: 29084133 PMCID: PMC5713245 DOI: 10.3390/ijms18112275] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/19/2017] [Accepted: 10/25/2017] [Indexed: 11/16/2022] Open
Abstract
Senescent cells display an increase in the secretion of growth factors, inflammatory cytokines and proteolytic enzymes, termed the "senescence-associated-secretory-phenotype" (SASP), playing a major role in many age-related diseases. The phenolic compounds present in extra-virgin olive oil are inhibitors of oxidative damage and have been reported to play a protective role in inflammation-related diseases. Particularly, hydroxytyrosol and oleuropein are the most abundant and more extensively studied. Pre-senescent human lung (MRC5) and neonatal human dermal (NHDF) fibroblasts were used as cellular model to evaluate the effect of chronic (4-6 weeks) treatment with 1 μM hydroxytyrosol (HT) or 10 μM oleuropein aglycone (OLE) on senescence/inflammation markers. Both phenols were effective in reducing β-galactosidase-positive cell number and p16 protein expression. In addition, senescence/inflammation markers such as IL-6 and metalloprotease secretion, and Ciclooxigenase type 2 (COX-2) and α-smooth-actin levels were reduced by phenol treatments. In NHDF, COX-2 expression, Nuclear Factor κ-light-chain-enhancer of activated B cells (NFκB) protein level and nuclear localization were augmented with culture senescence and decreased by OLE and HT treatment. Furthermore, the inflammatory effect of Tumor Necrosis Factor α (TNFα) exposure was almost completely abolished in OLE- and HT-pre-treated NHDF. Thus, the modulation of the senescence-associated inflammatory phenotype might be an important mechanism underlying the beneficial effects of olive oil phenols.
Collapse
|
191
|
Moretti M, Fraga DB, Rodrigues ALS. Preventive and therapeutic potential of ascorbic acid in neurodegenerative diseases. CNS Neurosci Ther 2017; 23:921-929. [PMID: 28980404 DOI: 10.1111/cns.12767] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 12/17/2022] Open
Abstract
In this review, we summarize the involvement of ascorbic acid in neurodegenerative diseases by presenting available evidence on the behavioral and biochemical effects of this compound in animal models of neurodegeneration as well as the use of ascorbic acid as a therapeutic approach to alleviate neurodegenerative progression in clinical studies. Ascorbate, a reduced form of vitamin C, has gained interest for its multiple functions and mechanisms of action, contributing to the homeostasis of normal tissues and organs as well as to tissue regeneration. In the brain, ascorbate exerts neuromodulatory functions and scavenges reactive oxygen species generated during synaptic activity and neuronal metabolism. These are important properties as redox imbalance and abnormal protein aggregation constitute central mechanisms implicated in the pathogenesis of neurodegenerative diseases, including Alzheimer's, Parkinson's, and Huntington's diseases, multiple sclerosis, and amyotrophic lateral sclerosis. Indeed, several studies have indicated an association between low serum ascorbate concentrations and neurodegeneration. Moreover, ascorbic acid is a suitable candidate for supplying either antioxidant defense or modulation of neuronal and astrocytic metabolism under neurodegenerative conditions. Ascorbic acid acts mainly by decreasing oxidative stress and reducing the formation of protein aggregates, which may contribute to the reduction of cognitive and/or motor impairments observed in neurodegenerative processes. Although several studies support a possible role of ascorbic acid administration against neurodegeneration, more researches are essential to substantiate the existing results and accelerate the knowledge in this field.
Collapse
Affiliation(s)
- Morgana Moretti
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daiane Bittencourt Fraga
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
192
|
Zhao Y, Cong L, Jaber V, Lukiw WJ. Microbiome-Derived Lipopolysaccharide Enriched in the Perinuclear Region of Alzheimer's Disease Brain. Front Immunol 2017; 8:1064. [PMID: 28928740 PMCID: PMC5591429 DOI: 10.3389/fimmu.2017.01064] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/16/2017] [Indexed: 12/16/2022] Open
Abstract
Abundant clinical, epidemiological, imaging, genetic, molecular, and pathophysiological data together indicate that there occur an unusual inflammatory reaction and a disruption of the innate-immune signaling system in Alzheimer’s disease (AD) brain. Despite many years of intense study, the origin and molecular mechanics of these AD-relevant pathogenic signals are still not well understood. Here, we provide evidence that an intensely pro-inflammatory bacterial lipopolysaccharide (LPS), part of a complex mixture of pro-inflammatory neurotoxins arising from abundant Gram-negative bacilli of the human gastrointestinal (GI) tract, are abundant in AD-affected brain neocortex and hippocampus. For the first time, we provide evidence that LPS immunohistochemical signals appear to aggregate in clumps in the parenchyma in control brains, and in AD, about 75% of anti-LPS signals were clustered around the periphery of DAPI-stained nuclei. As LPS is an abundant secretory product of Gram-negative bacilli resident in the human GI-tract, these observations suggest (i) that a major source of pro-inflammatory signals in AD brain may originate from internally derived noxious exudates of the GI-tract microbiome; (ii) that due to aging, vascular deficits or degenerative disease these neurotoxic molecules may “leak” into the systemic circulation, cerebral vasculature, and on into the brain; and (iii) that this internal source of microbiome-derived neurotoxins may play a particularly strong role in shaping the human immune system and contributing to neural degeneration, particularly in the aging CNS. This “Perspectives” paper will further highlight some very recent developments that implicate GI-tract microbiome-derived LPS as an important contributor to inflammatory-neurodegeneration in the AD brain.
Collapse
Affiliation(s)
- Yuhai Zhao
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Anatomy and Cell Biology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Lin Cong
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Neurology, Shengjing Hospital, China Medical University, Heping District, Shenyang, China
| | - Vivian Jaber
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Walter J Lukiw
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Neurology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Ophthalmology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
193
|
Tzeng NS, Chang HA, Chung CH, Lin FH, Yeh CB, Huang SY, Chang CC, Lu RB, Kao YC, Yeh HW, Chiang WS, Chien WC. Risk of psychiatric disorders in Guillain-Barre syndrome: A nationwide, population-based, cohort study. J Neurol Sci 2017; 381:88-94. [PMID: 28991722 DOI: 10.1016/j.jns.2017.08.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/25/2017] [Accepted: 08/16/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND Guillain-Barre syndrome (GBS) is a rare immune-related neurological disorder with high mortality and morbidity, but the comorbid psychiatric disorders garnered little attention in the GBS patients. This study aimed to investigate the association between GBS and the risk of developing psychiatric disorders. METHODS A total of 18,192 enrolled patients, with 4548 study subjects who had suffered GBS, and 13,644 controls matched for gender and age, from the Inpatient Dataset of 2000-2013 in Taiwan, and selected from the National Health Insurance Research Database (NHIRD). After adjusting for confounding factors, Cox proportional hazards analysis was used to compare the risk of developing psychiatric disorders during the 13years of follow-up. RESULTS Of the study subjects, 471 (10.35%) developed psychiatric disorders when compared to 1023 (7.50%) in the control group. Fine and Gray's competing risk model analysis revealed that the study subjects were more likely to develop psychiatric disorders (crude hazard ratio [HR]: 4.281 (95% CI=3.819-4.798, p<0.001). After adjusting for gender, age, monthly income, urbanization level, geographic region, and comorbidities, the adjusted HR was 4.320 (95% CI=3.852-4.842, p<0.001). Dementia, depressive disorders, sleep disorders, and psychotic disorders predominate in these psychiatric disorders. Mechanical ventilation and hemodialysis are associated with a lower risk of dementia when compared to the control groups. CONCLUSIONS Patients who suffered from GBS had a higher risk of developing psychiatric disorders, and this finding should act as a reminder to the clinicians that a regular psychiatric follow-up might well be needed for those patients.
Collapse
Affiliation(s)
- Nian-Sheng Tzeng
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC; Student Counseling Center, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hsin-An Chang
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC; Student Counseling Center, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chi-Hsiang Chung
- Taiwanese Injury Prevention and Safety Promotion Association, Taipei, Taiwan, ROC; School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Fu-Huang Lin
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chin-Bin Yeh
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - San-Yuan Huang
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chuan-Chia Chang
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Ru-Band Lu
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Division of Clinical Psychology, Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC; Department of Psychiatry, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC; Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC; Department of Psychiatry, National Cheng Kung University Hospital, Tainan, Taiwan, ROC; Center for Neuropsychiatric Research, National Health Research Institute, Zhunan, Miaoli County, Taiwan, ROC
| | - Yu-Chen Kao
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Psychiatry, Tri-Service General Hospital, Song-Shan Branch, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hui-Wen Yeh
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC; Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsin-Chu, Taiwan, ROC; Department of Nursing, Tri-Service General Hospital, School of Nursing, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Nursing, Kang-Ning University (Taipei Campus), Taipei, Taiwan, ROC
| | - Wei-Shan Chiang
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC; Department and Institute of Mathematics, Tamkang University, New Taipei City, Taiwan, ROC
| | - Wu-Chien Chien
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
194
|
Le Page A, Garneau H, Dupuis G, Frost EH, Larbi A, Witkowski JM, Pawelec G, Fülöp T. Differential Phenotypes of Myeloid-Derived Suppressor and T Regulatory Cells and Cytokine Levels in Amnestic Mild Cognitive Impairment Subjects Compared to Mild Alzheimer Diseased Patients. Front Immunol 2017; 8:783. [PMID: 28736551 PMCID: PMC5500623 DOI: 10.3389/fimmu.2017.00783] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/21/2017] [Indexed: 12/26/2022] Open
Abstract
Alzheimer disease (AD) is the most prevalent form of dementia although the underlying cause(s) remains unknown at this time. However, neuroinflammation is believed to play an important role and suspected contributing immune parameters can be revealed in studies comparing patients at the stage of amnestic mild cognitive impairment (aMCI) to healthy age-matched individuals. A network of immune regulatory cells including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) maintains immune homeostasis but there are very few data on the role of these cells in AD. Here, we investigated the presence of these cells in the blood of subjects with aMCI and mild AD (mAD) in comparison with healthy age-matched controls. We also quantitated several pro- and anti-inflammatory cytokines in sera which can influence the development and activation of these cells. We found significantly higher levels of MDSCs and Tregs in aMCI but not in mAD patients, as well as higher serum IL-1β levels. Stratifying the subjects based on CMV serostatus that is known to influence multiple immune parameters showed an absence of differences between aMCI subjects compared to mAD patients and healthy controls. We suggest that the increase in MDSCs and Tregs number in aMCI subjects may have a beneficial role in modulating inflammatory processes. However, this protective mechanism may have failed in mAD patients, allowing progression of the disease. This working hypothesis obviously requires testing in future studies.
Collapse
Affiliation(s)
- Aurélie Le Page
- Faculty of Medicine and Health Sciences, Research Center on Aging, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Hugo Garneau
- Faculty of Medicine and Health Sciences, Research Center on Aging, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Gilles Dupuis
- Faculty of Medicine and Health Sciences, Department of Biochemistry, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Eric H Frost
- Faculty of Medicine and Health Sciences, Department of Infectious Diseases and Microbiology, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Anis Larbi
- ASTAR, Singapore Immunology Network, Singapore, Singapore
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Graham Pawelec
- Department of Internal Medicine II, Center for Medical Research University of Tübingen, Tübingen, Germany.,Health Sciences North Research Institute, Sudbury, ON, Canada
| | - Tamàs Fülöp
- Faculty of Medicine and Health Sciences, Research Center on Aging, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
195
|
Xia L, Xie X, Liu Y, Luo X. Peripheral Blood Monocyte Tolerance Alleviates Intraperitoneal Lipopolysaccharides-Induced Neuroinflammation in Rats Via Upregulating the CD200R Expression. Neurochem Res 2017; 42:3019-3032. [DOI: 10.1007/s11064-017-2334-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/01/2017] [Accepted: 06/16/2017] [Indexed: 01/17/2023]
|