1
|
Sarkar S, Porel P, Kosey S, Aran KR. Diverse role of S100 calcium-binding protein B in alzheimer's disease: pathological mechanisms and therapeutic implications. Inflammopharmacology 2025; 33:1803-1816. [PMID: 40057929 DOI: 10.1007/s10787-025-01697-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/01/2025] [Indexed: 04/13/2025]
Abstract
S100 calcium-binding protein B, a member of the S100 protein family, plays an important role in the pathogenesis of Alzheimer's disease. Alzheimer's disease, a neurodegenerative disorder, is characterized by amyloid-beta plaques, neurofibrillary tangles, progressive dementia, and severe neuroinflammation. S100 calcium-binding protein B, predominantly secreted by astrocytes, exhibits a dual role in Alzheimer's disease, where at low (nanomolar) concentrations, it exhibits neurotrophic and neuroprotective effects and enhances synaptic plasticity, while at higher concentrations, it contributes to neuroinflammation and neuronal damage. In addition to its pathological roles in Alzheimer's disease, S100 calcium-binding protein B is also considered a potential biomarker, as increased levels correlate with cognitive decline and disease progression in cerebrospinal fluid. Targeting S100 calcium-binding protein B and/or its interaction with the receptor for advanced glycation end-products seems to be a potential target for therapeutic intervention. The development of multiple treatment approaches, such as pharmacological inhibitors, immunotherapy, and modulation of S100 calcium-binding protein B / receptor for advanced glycation end-products signalling pathways, might help to reduce neuroinflammation and amyloid-beta deposition effectively. This review aims to provide an overview of the role of S100 calcium-binding protein B in Alzheimer's disease and to explore its potential as a treatment target, well-grounded in its dual nature. Understanding S100 calcium-binding protein B's involvement in the pathogenesis of Alzheimer's disease may advance its application as a biomarker and help in the development of new treatment strategies, ultimately improving patients' quality of life.
Collapse
Affiliation(s)
- Sampriti Sarkar
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Pratyush Porel
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Sourabh Kosey
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Khadga Raj Aran
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
2
|
Yu H, Ren K, Jin Y, Zhang L, Liu H, Huang Z, Zhang Z, Chen X, Yang Y, Wei Z. Mitochondrial DAMPs: Key mediators in neuroinflammation and neurodegenerative disease pathogenesis. Neuropharmacology 2025; 264:110217. [PMID: 39557152 DOI: 10.1016/j.neuropharm.2024.110217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are increasingly linked to mitochondrial dysfunction and neuroinflammation. Central to this link are mitochondrial damage-associated molecular patterns (mtDAMPs), including mitochondrial DNA, ATP, and reactive oxygen species, released during mitochondrial stress or damage. These mtDAMPs activate inflammatory pathways, such as the NLRP3 inflammasome and cGAS-STING, contributing to the progression of neurodegenerative diseases. This review delves into the mechanisms by which mtDAMPs drive neuroinflammation and discusses potential therapeutic strategies targeting these pathways to mitigate neurodegeneration. Additionally, it explores the cross-talk between mitochondria and the immune system, highlighting the complex interplay that exacerbates neuronal damage. Understanding the role of mtDAMPs could pave the way for novel treatments aimed at modulating neuroinflammation and slowing disease progression, ultimately improving patient outcome.
Collapse
Affiliation(s)
- Haihan Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Li Zhang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Hui Liu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Zhen Huang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Ziheng Zhang
- College of Life Sciences, Xinjiang University, Urumqi, Xinjiang, 830046, PR China
| | - Xing Chen
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
3
|
Deng Z, Lee A, Lin T, Taneja S, Kowdley D, Leung JH, Hill M, Tao T, Fitzgerald J, Yu L, Blakeslee JJ, Townsend K, Weil ZM, Parquette JR, Ziouzenkova O. Amino Acid Compound 2 (AAC2) Treatment Counteracts Insulin-Induced Synaptic Gene Expression and Seizure-Related Mortality in a Mouse Model of Alzheimer's Disease. Int J Mol Sci 2024; 25:11689. [PMID: 39519239 PMCID: PMC11546384 DOI: 10.3390/ijms252111689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Diabetes is a major risk factor for Alzheimer's disease (AD). Amino acid compound 2 (AAC2) improves glycemic and cognitive functions in diabetic mouse models through mechanisms distinct from insulin. Our goal was to compare the effects of AAC2, insulin, and their nanofiber-forming combination on early asymptomatic AD pathogenesis in APP/PS1 mice. Insulin, but not AAC2 or the combination treatment (administered intraperitoneally every 48 h for 120 days), increased seizure-related mortality, altered the brain fat-to-lean mass ratio, and improved specific cognitive functions in APP/PS1 mice. NanoString and pathway analysis of cerebral gene expression revealed dysregulated synaptic mechanisms, with upregulation of Bdnf and downregulation of Slc1a6 in insulin-treated mice, correlating with insulin-induced seizures. In contrast, AAC2 promoted the expression of Syn2 and Syp synaptic genes, preserved brain composition, and improved survival. The combination of AAC2 and insulin counteracted free insulin's effects. None of the treatments influenced canonical amyloidogenic pathways. This study highlights AAC2's potential in regulating synaptic gene expression in AD and insulin-induced contexts related to seizure activity.
Collapse
Affiliation(s)
- Zhijie Deng
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; (Z.D.); or (A.L.); (D.K.); (J.H.L.); (M.H.)
| | - Aejin Lee
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; (Z.D.); or (A.L.); (D.K.); (J.H.L.); (M.H.)
- Department of Food and Nutrition, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si 17058, Gyeonggi-do, Republic of Korea
| | - Tao Lin
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; (T.L.); (S.T.); (J.R.P.)
| | - Sagarika Taneja
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; (T.L.); (S.T.); (J.R.P.)
| | - Devan Kowdley
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; (Z.D.); or (A.L.); (D.K.); (J.H.L.); (M.H.)
| | - Jacob H. Leung
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; (Z.D.); or (A.L.); (D.K.); (J.H.L.); (M.H.)
| | - Marykate Hill
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; (Z.D.); or (A.L.); (D.K.); (J.H.L.); (M.H.)
| | - Tianyi Tao
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (T.T.); (K.T.)
| | - Julie Fitzgerald
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; (J.F.); (Z.M.W.)
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210, USA;
| | - Joshua J. Blakeslee
- Department of Horticulture and Crop Science, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Columbus, OH 43210, USA;
| | - Kristy Townsend
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (T.T.); (K.T.)
| | - Zachary M. Weil
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA; (J.F.); (Z.M.W.)
- Department of Neuroscience, WVU Rockefeller Neuroscience Institute, West Virginia University, Biomedical Research Center (BMRC), Morgantown, WV 26506, USA
| | - Jon R. Parquette
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA; (T.L.); (S.T.); (J.R.P.)
| | - Ouliana Ziouzenkova
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; (Z.D.); or (A.L.); (D.K.); (J.H.L.); (M.H.)
| |
Collapse
|
4
|
Lahiri DK, Maloney B, Wang R, White FA, Sambamurti K, Greig NH, Counts SE. The seeds of its regulation: Natural antisense transcripts as single-gene control switches in neurodegenerative disorders. Ageing Res Rev 2024; 99:102336. [PMID: 38740308 PMCID: PMC11492926 DOI: 10.1016/j.arr.2024.102336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Several proteins play critical roles in vulnerability or resistance to neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and frontotemporal dementia (FTD). Regulation of these proteins is critical to maintaining healthy neurohomeostasis. In addition to transcription factors regulating gene transcription and microRNAs regulating mRNA translation, natural antisense transcripts (NATs) regulate mRNA levels, splicing, and translation. NATs' roles are significant in regulating key protein-coding genes associated with neurodegenerative disorders. Elucidating the functions of these NATs could prove useful in treating or preventing diseases. NAT activity is not restricted to mRNA translation; it can also regulate DNA (de)methylation and other gene expression steps. NATs are noncoding RNAs (ncRNAs) encoded by DNA sequences overlapping the pertinent protein genes. These NATs have complex structures, including introns and exons, and therefore bind their target genes, precursor mRNAs (pre-mRNAs), and mature RNAs. They can occur at the 5'- or 3'-ends of a mRNA-coding sequence or internally to a parent gene. NATs can downregulate translation, e.g., microtubule-associated protein tau (MAPT) antisense-1 gene (MAPT-AS1), or upregulate translation, e.g., β-Amyloid site Cleaving Enzyme 1 (BACE1) antisense gene (BACE1-AS). Regulation of NATs can parallel pathogenesis, wherein a "pathogenic" NAT (e.g., BACE1-AS) is upregulated under pathogenic conditions, while a "protective" NAT (e.g., MAPT-AS1) is downregulated under pathogenic conditions. As a relatively underexplored endogenous control mechanism of protein expression, NATs may present novel mechanistic targets to prevent or ameliorate aging-related disorders.
Collapse
Affiliation(s)
- Debomoy K Lahiri
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Bryan Maloney
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ruizhi Wang
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kumar Sambamurti
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Scott E Counts
- Departments of Translational Neuroscience and Family Medicine, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
5
|
Cho SB. Comorbidity Genes of Alzheimer's Disease and Type 2 Diabetes Associated with Memory and Cognitive Function. Int J Mol Sci 2024; 25:2211. [PMID: 38396891 PMCID: PMC10889845 DOI: 10.3390/ijms25042211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/02/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are comorbidities that result from the sharing of common genes. The molecular background of comorbidities can provide clues for the development of treatment and management strategies. Here, the common genes involved in the development of the two diseases and in memory and cognitive function are reviewed. Network clustering based on protein-protein interaction network identified tightly connected gene clusters that have an impact on memory and cognition among the comorbidity genes of AD and T2DM. Genes with functional implications were intensively reviewed and relevant evidence summarized. Gene information will be useful in the discovery of biomarkers and the identification of tentative therapeutic targets for AD and T2DM.
Collapse
Affiliation(s)
- Seong Beom Cho
- Department of Biomedical Informatics, College of Medicine, Gachon University, 38-13, Dokgeom-ro 3 Street, Namdon-gu, Incheon 21565, Republic of Korea
| |
Collapse
|
6
|
Coelho R, De Benedictis CA, Sauer AK, Figueira AJ, Faustino H, Grabrucker AM, Gomes CM. Secondary Modification of S100B Influences Anti Amyloid-β Aggregation Activity and Alzheimer's Disease Pathology. Int J Mol Sci 2024; 25:1787. [PMID: 38339064 PMCID: PMC10855146 DOI: 10.3390/ijms25031787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Proteinaceous aggregates accumulate in neurodegenerative diseases such as Alzheimer's Disease (AD), inducing cellular defense mechanisms and altering the redox status. S100 pro-inflammatory cytokines, particularly S100B, are activated during AD, but recent findings reveal an unconventional molecular chaperone role for S100B in hindering Aβ aggregation and toxicity. This suggests a potential protective role for S100B at the onset of Aβ proteotoxicity, occurring in a complex biochemical environment prone to oxidative damage. Herein, we report an investigation in which extracellular oxidative conditions are mimicked to test if the susceptibility of S100B to oxidation influences its protective activities. Resorting to mild oxidation of S100B, we observed methionine oxidation as inferred from mass spectrometry, but no cysteine-mediated crosslinking. Structural analysis showed that the folding, structure, and stability of oxidized S100B were not affected, and nor was its quaternary structure. However, studies on Aβ aggregation kinetics indicated that oxidized S100B was more effective in preventing aggregation, potentially linked to the oxidation of Met residues within the S100:Aβ binding cleft that favors interactions. Using a cell culture model to analyze the S100B functions in a highly oxidative milieu, as in AD, we observed that Aβ toxicity is rescued by the co-administration of oxidized S100B to a greater extent than by S100B. Additionally, results suggest a disrupted positive feedback loop involving S100B which is caused by its oxidation, leading to the downstream regulation of IL-17 and IFN-α2 expression as mediated by S100B.
Collapse
Affiliation(s)
- Romina Coelho
- BioISI—Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal; (R.C.); (A.J.F.)
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Chiara A. De Benedictis
- Cellular Neurobiology and Neuro-Nanotechnology Laboratory, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland; (C.A.D.B.); (A.K.S.)
- Bernal Institute, University of Limerick, V94PH61 Limerick, Ireland
| | - Ann Katrin Sauer
- Cellular Neurobiology and Neuro-Nanotechnology Laboratory, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland; (C.A.D.B.); (A.K.S.)
- Bernal Institute, University of Limerick, V94PH61 Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94PH61 Limerick, Ireland
| | - António J. Figueira
- BioISI—Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal; (R.C.); (A.J.F.)
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Hélio Faustino
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
- Association BLC3—Technology and Innovation Campus, Centre Bio R&D Unit, Oliveira do Hospital, Rua Nossa Senhora da Conceição No. 2, 3405-155 Coimbra, Portugal
| | - Andreas M. Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology Laboratory, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland; (C.A.D.B.); (A.K.S.)
- Bernal Institute, University of Limerick, V94PH61 Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94PH61 Limerick, Ireland
| | - Cláudio M. Gomes
- BioISI—Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal; (R.C.); (A.J.F.)
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| |
Collapse
|
7
|
Luo EY, Sugimura RR. Taming microglia: the promise of engineered microglia in treating neurological diseases. J Neuroinflammation 2024; 21:19. [PMID: 38212785 PMCID: PMC10785527 DOI: 10.1186/s12974-024-03015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Microglia, the CNS-resident immune cells, are implicated in many neurological diseases. Nearly one in six of the world's population suffers from neurological disorders, encompassing neurodegenerative and neuroautoimmune diseases, most with dysregulated neuroinflammation involved. Activated microglia become phagocytotic and secret various immune molecules, which are mediators of the brain immune microenvironment. Given their ability to penetrate through the blood-brain barrier in the neuroinflammatory context and their close interaction with neurons and other glial cells, microglia are potential therapeutic delivery vehicles and modulators of neuronal activity. Re-engineering microglia to treat neurological diseases is, thus, increasingly gaining attention. By altering gene expression, re-programmed microglia can be utilized to deliver therapeutics to targeted sites and control neuroinflammation in various neuroinflammatory diseases. This review addresses the current development in microglial engineering, including genetic targeting and therapeutic modulation. Furthermore, we discuss limitations to the genetic engineering techniques and models used to test the functionality of re-engineered microglia, including cell culture and animal models. Finally, we will discuss future directions for the application of engineered microglia in treating neurological diseases.
Collapse
Affiliation(s)
- Echo Yongqi Luo
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Pokfulam, Hong Kong
| | - Rio Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
8
|
Bogolepova AN, Mkhitaryan EA, Levin OS. [Cognitive impairment in cerebrovascular diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:12-16. [PMID: 38696146 DOI: 10.17116/jnevro202412404212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Cognitive impairment, which is highly prevalent, especially among older people, leads to a decrease in the quality of life of patients, impairment of daily activities, and an increased risk of dementia and mortality. Currently, much attention is paid to mild cognitive impairment. The article discusses diagnostic criteria and possible clinical variants of this syndrome. Given the high rate of progression of mild cognitive impairment to dementia, it is necessary to identify risk groups and carry out therapeutic preventive measures. Correction of potentially modifiable risk factors is considered as a promising direction of therapy. Sufficient physical and mental activity, proper diet, normalization of sleep, visual acuity and hearing are necessary. Preventing stroke and controlling vascular risk factors may reduce the risk of mild cognitive impairment progressing to dementia.
Collapse
Affiliation(s)
- A N Bogolepova
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain and Neurotechnologies, Moscow, Russia
| | - E A Mkhitaryan
- Pirogov Russian National Research Medical University, Moscow, Russia
- Russian Gerontology Research and Clinical Centre - Pirogov Russian National Research Medical University, Moscow, Russia
| | - O S Levin
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
- S.P. Botkin City Clinical Hospital, Moscow, Russia
| |
Collapse
|
9
|
Eremin DV, Kondaurova EM, Rodnyy AY, Molobekova CA, Kudlay DA, Naumenko VS. Serotonin Receptors as a Potential Target in the Treatment of Alzheimer's Disease. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:2023-2042. [PMID: 38462447 DOI: 10.1134/s0006297923120064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 03/12/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide that has an increasing impact on aging societies. Besides its critical role in the control of various physiological functions and behavior, brain serotonin (5-HT) system is involved in the regulation of migration, proliferation, differentiation, maturation, and programmed death of neurons. At the same time, a growing body of evidence indicates the involvement of 5-HT neurotransmission in the formation of insoluble aggregates of β-amyloid and tau protein, the main histopathological signs of AD. The review describes the role of various 5-HT receptors and intracellular signaling cascades induced by them in the pathological processes leading to the development of AD, first of all, in protein aggregation. Changes in the functioning of certain types of 5-HT receptors or associated intracellular signaling mediators prevent accumulation of β-amyloid plaques and tau protein neurofibrillary tangles. Based on the experimental data, it can be suggested that the use of 5-HT receptors as new drug targets will not only improve cognitive performance in AD, but will be also important in treating the causes of AD-related dementia.
Collapse
Affiliation(s)
- Dmitrii V Eremin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| | - Elena M Kondaurova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Aleksander Ya Rodnyy
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Camilla A Molobekova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Dmitrii A Kudlay
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, 119991, Russia
| | - Vladimir S Naumenko
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| |
Collapse
|
10
|
Moreira GG, Gomes CM. Tau liquid-liquid phase separation is modulated by the Ca 2+ -switched chaperone activity of the S100B protein. J Neurochem 2023; 166:76-86. [PMID: 36621842 DOI: 10.1111/jnc.15756] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/03/2022] [Accepted: 12/19/2022] [Indexed: 01/10/2023]
Abstract
Aggregation of the microtubule-associated protein tau is implicated in several neurodegenerative tauopathies including Alzheimer's disease (AD). Recent studies evidenced tau liquid-liquid phase separation (LLPS) into droplets as an early event in tau pathogenesis with the potential to enhance aggregation. Tauopathies like AD are accompanied by sustained neuroinflammation and the release of alarmins at early stages of inflammatory responses encompass protective functions. The Ca2+ -binding S100B protein is an alarmin augmented in AD that was recently implicated as a proteostasis regulator acting as a chaperone-type protein, inhibiting aggregation and toxicity through interactions of amyloidogenic clients with a regulatory surface exposed upon Ca2+ -binding. Here we expand the regulatory functions of S100B over protein condensation phenomena by reporting its Ca2+ -dependent activity as a modulator of tau LLPS induced by crowding agents (PEG) and metal ions (Zn2+ ). We observe that apo S100B has a negligible effect on PEG-induced tau demixing but that Ca2+ -bound S100B prevents demixing, resulting in a shift of the phase diagram boundary to higher crowding concentrations. Also, while incubation with apo S100B does not compromise tau LLPS, addition of Ca2+ results in a sharp decrease in turbidity, indicating that interactions with S100B-Ca2+ promote transition of tau to the mixed phase. Further, electrophoretic analysis and FLIM-FRET studies revealed that S100B incorporates into tau liquid droplets, suggesting an important stabilizing and chaperoning role contributing to minimize toxic tau aggregates. Resorting to Alexa488-labeled tau we observed that S100B-Ca2+ reduces the formation of tau fluorescent droplets, without compromising liquid-like behavior and droplet fusion events. The Zn2+ -binding properties of S100B also contribute to regulate Zn2+ -promoted tau LLPS as droplets are decreased by Zn2+ buffering by S100B, in addition to the Ca2+ -triggered interactions with tau. Altogether this work uncovers the versatility of S100B as a proteostasis regulator acting on protein condensation phenomena of relevance across the neurodegeneration continuum.
Collapse
Affiliation(s)
- Guilherme G Moreira
- BioISI-Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Cláudio M Gomes
- BioISI-Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
11
|
Gerber K, Iriarte E, Sierra CJ. Multidimensional Frailty and Traumatic Brain Injury among Older Adults: A Literature Review. INVESTIGACION Y EDUCACION EN ENFERMERIA 2023; 41:e02. [PMID: 38589320 PMCID: PMC10599699 DOI: 10.17533/udea.iee.v41n2e02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 05/25/2023] [Indexed: 04/10/2024]
Abstract
Background Numerous health conditions in the older adult population can be attributed to falls, including traumatic brain injury (TBI), which can lead to devastating short and long-term sequelae. Older adults are also more likely to experience frailty, which encompasses physical, psychological, and social deficits that may lead to adverse health outcomes. Our literature review synthesizes current evidence for understanding frailty in the context of TBI among older adults using the Integral Model of Frailty as a framework. Content Synthesis A total of 32 articles were identified, and 9 articles were included. The results of this review indicate that outcomes resulting from TBI are closely linked to the physical, psychological, and social domains of frailty. Conclusions A small amount of literature currently examines frailty in the context of TBI among older adults. Using the Integral Model of Frailty to understand frailty in the context of TBI can help clinicians anticipate patient outcomes and improve care plans. We emphasize the need for a greater understanding of TBI concerning frailty to improve health outcomes among older adult patients.
Collapse
Affiliation(s)
- Kathryn Gerber
- RN. Ph.D. School of Nursing and Health Studies, University of Miami, Coral Gables, Florida, U.S.
| | - Evelyn Iriarte
- Ph.D, MSN, RN. Adjunct Instructor at the School of Nursing, Pontificia Universidad Catolica de Chile, Santiago, Chile. Postdoctoral Fellow at the University of Colorado College of Nursing, Aurora, Colorado, U.S., and Young Researcher at Millennium Institute for Care Research, MICARE, Santiago, Chile.
| | - Carmen Josefa Sierra
- DNP, RN, CCTN. School of Nursing and Health Studies, University of Miami, Coral Gables, Florida, U.S.
| |
Collapse
|
12
|
Khamis M, Din NSE, Nada MA, Afifi HEDM. Serum protein S-100B as a novel biomarker of diagnosis and prognosis of childhood epilepsy. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2023. [DOI: 10.1186/s41983-023-00605-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Abstract
Background
Elevated levels of S-100B in serum are increasingly considered a potential biochemical marker of nervous system damage. To our knowledge, limited number of research studies have tested the serum S-100B protein levels in children with epilepsy. The objective of our study is to measure the serum levels of S-100B protein in pediatric cases with epilepsy.
Results
The mean serum concentration of S-100B protein was 0.135 ± 0.014 mg/L in the patient group and 0.082 ± 0.018 mg/L in the control group. The patients showed significantly high S-100B protein levels compared with healthy controls (P < 0.001).
Conclusion
Our data suggest that increased S-100B protein levels in the serum potentially indicate neuronal damage in the brains of children with epilepsy.
Collapse
|
13
|
Mercado-Gómez OF, Arriaga-Ávila VS, Vega-García A, Sánchez-Hernández J, Jiménez A, Organista-Juárez D, Guzmán-Ruiz MA, Guevara-Guzmán R. Cellular and Molecular Mechanisms of Neuroinflammation in Drug-Resistant Epilepsy. PHARMACORESISTANCE IN EPILEPSY 2023:131-156. [DOI: 10.1007/978-3-031-36526-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
14
|
Diaz-Amarilla P, Arredondo F, Dapueto R, Boix V, Carvalho D, Santi MD, Vasilskis E, Mesquita-Ribeiro R, Dajas-Bailador F, Abin-Carriquiry JA, Engler H, Savio E. Isolation and characterization of neurotoxic astrocytes derived from adult triple transgenic Alzheimer's disease mice. Neurochem Int 2022; 159:105403. [PMID: 35853553 DOI: 10.1016/j.neuint.2022.105403] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/02/2022] [Accepted: 07/09/2022] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease has been considered mostly as a neuronal pathology, although increasing evidence suggests that glial cells might play a key role in the disease onset and progression. In this sense, astrocytes, with their central role in neuronal metabolism and function, are of great interest for increasing our understanding of the disease. Thus, exploring the morphological and functional changes suffered by astrocytes along the course of this disorder has great therapeutic and diagnostic potential. In this work we isolated and cultivated astrocytes from symptomatic 9-10-months-old adult 3xTg-AD mice, with the aim of characterizing their phenotype and exploring their pathogenic potential. These "old" astrocytes occurring in the 3xTg-AD mouse model of Alzheimer's Disease presented high proliferation rate and differential expression of astrocytic markers compared with controls. They were neurotoxic to primary neuronal cultures both, in neuronal-astrocyte co-cultures and when their conditioned media (ACM) was added into neuronal cultures. ACM caused neuronal GSK3β activation, changes in cytochrome c pattern, and increased caspase 3 activity, suggesting intrinsic apoptotic pathway activation. Exposure of neurons to ACM caused different subcellular responses. ACM application to the somato-dendritic domain in compartmentalised microfluidic chambers caused degeneration both locally in soma/dendrites and distally in axons. However, exposure of axons to ACM did not affect somato-dendritic nor axonal integrity. We propose that this newly described old 3xTg-AD neurotoxic astrocytic population can contribute towards the mechanistic understanding of the disease and shed light on new therapeutical opportunities.
Collapse
Affiliation(s)
- Pablo Diaz-Amarilla
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Florencia Arredondo
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay; Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay.
| | - Rosina Dapueto
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Victoria Boix
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - Diego Carvalho
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - María Daniela Santi
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Elena Vasilskis
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay
| | - Raquel Mesquita-Ribeiro
- School of Life Sciences, Medical School Building, University of Nottingham, NG7 2UH, Nottingham, UK
| | - Federico Dajas-Bailador
- School of Life Sciences, Medical School Building, University of Nottingham, NG7 2UH, Nottingham, UK
| | - Juan Andrés Abin-Carriquiry
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - Henry Engler
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay.
| | - Eduardo Savio
- Area I+D Biomédica, Centro Uruguayo de Imagenología Molecular, 11600, Montevideo, Uruguay.
| |
Collapse
|
15
|
Gerber KS, Alvarez G, Alamian A, Behar-Zusman V, Downs CA. Biomarkers of Neuroinflammation in Traumatic Brain Injury. Clin Nurs Res 2022; 31:1203-1218. [PMID: 35770330 DOI: 10.1177/10547738221107081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Traumatic brain injury (TBI) is characterized by neuroinflammation and structural damage leading to symptoms and altered brain function. Biomarkers are useful in understanding neuroinflammation and correlations with TBI sequalae. The purpose of this paper is to identify and discuss biomarkers of neuroinflammation used to study TBI and its sequalae. A systematic review was conducted using PubMed, CINAHL, Embase, and Web of Science. A total of 350 articles met criteria; 70 used biomarkers. PRISMA criteria were used for Quality Assessment. Articles included reviews (n = 17), case-control (n = 25), cross-sectional (n = 25) studies, and randomized controlled trials (n = 3). Twenty-seven biomarkers were identified, including inflammasomes, cytokines, neuropeptides, complement complexes, miRNA and exosomes, and glial cell-specific proteins. Biomarkers aid in predicting morbidity and mortality and advance our understanding of neuroinflammation in TBI. This systematic review advances our understanding of the neuroinflammatory response to better enable nurses and clinicians to provide informed care of TBI patients.
Collapse
Affiliation(s)
- Kathryn S Gerber
- University of Miami School of Nursing and Health Studies, Coral Gables, FL, USA
| | - Gema Alvarez
- University of Miami Miller School of Medicine, FL, USA
| | - Arsham Alamian
- University of Miami School of Nursing and Health Studies, Coral Gables, FL, USA
| | | | - Charles A Downs
- University of Miami School of Nursing and Health Studies, Coral Gables, FL, USA
| |
Collapse
|
16
|
Zaręba-Kozioł M, Burdukiewicz M, Wysłouch-Cieszyńska A. Intracellular Protein S-Nitrosylation—A Cells Response to Extracellular S100B and RAGE Receptor. Biomolecules 2022; 12:biom12050613. [PMID: 35625541 PMCID: PMC9138530 DOI: 10.3390/biom12050613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/18/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
Human S100B is a small, multifunctional protein. Its activity, inside and outside cells, contributes to the biology of the brain, muscle, skin, and adipocyte tissues. Overexpression of S100B occurs in Down Syndrome, Alzheimer’s disease, Creutzfeldt–Jakob disease, schizophrenia, multiple sclerosis, brain tumors, epilepsy, melanoma, myocardial infarction, muscle disorders, and sarcopenia. Modulating the activities of S100B, related to human diseases, without disturbing its physiological functions, is vital for drug and therapy design. This work focuses on the extracellular activity of S100B and one of its receptors, the Receptor for Advanced Glycation End products (RAGE). The functional outcome of extracellular S100B, partially, depends on the activation of intracellular signaling pathways. Here, we used Biotin Switch Technique enrichment and mass-spectrometry-based proteomics to show that the appearance of the S100B protein in the extracellular milieu of the mammalian Chinese Hamster Ovary (CHO) cells, and expression of the membrane-bound RAGE receptor, lead to changes in the intracellular S-nitrosylation of, at least, more than a hundred proteins. Treatment of the wild-type CHO cells with nanomolar or micromolar concentrations of extracellular S100B modulates the sets of S-nitrosylation targets inside cells. The cellular S-nitrosome is tuned differently, depending on the presence or absence of stable RAGE receptor expression. The presented results are a proof-of-concept study, suggesting that S-nitrosylation, like other post-translational modifications, should be considered in future research, and in developing tailored therapies for S100B and RAGE receptor-related diseases.
Collapse
Affiliation(s)
- Monika Zaręba-Kozioł
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland;
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Science, Pasteura 3, 02-093 Warsaw, Poland
| | - Michał Burdukiewicz
- Clinical Research Centre, Medical University of Białystok, Kilińskiego 1, 15-369 Białystok, Poland;
| | - Aleksandra Wysłouch-Cieszyńska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland;
- Correspondence:
| |
Collapse
|
17
|
Antioxidants in Alzheimer's Disease: Current Therapeutic Significance and Future Prospects. BIOLOGY 2022; 11:biology11020212. [PMID: 35205079 PMCID: PMC8869589 DOI: 10.3390/biology11020212] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 01/27/2023]
Abstract
Alzheimer's disease (AD) rate is accelerating with the increasing aging of the world's population. The World Health Organization (WHO) stated AD as a global health priority. According to the WHO report, around 82 million people in 2030 and 152 million in 2050 will develop dementia (AD contributes 60% to 70% of cases), considering the current scenario. AD is the most common neurodegenerative disease, intensifying impairments in cognition, behavior, and memory. Histopathological AD variations include extracellular senile plaques' formation, tangling of intracellular neurofibrils, and synaptic and neuronal loss in the brain. Multiple evidence directly indicates that oxidative stress participates in an early phase of AD before cytopathology. Moreover, oxidative stress is induced by almost all misfolded protein lumps like α-synuclein, amyloid-β, and others. Oxidative stress plays a crucial role in activating and causing various cell signaling pathways that result in lesion formations of toxic substances, which foster the development of the disease. Antioxidants are widely preferred to combat oxidative stress, and those derived from natural sources, which are often incorporated into dietary habits, can play an important role in delaying the onset as well as reducing the progression of AD. However, this approach has not been extensively explored yet. Moreover, there has been growing evidence that a combination of antioxidants in conjugation with a nutrient-rich diet might be more effective in tackling AD pathogenesis. Thus, considering the above-stated fact, this comprehensive review aims to elaborate the basics of AD and antioxidants, including the vitality of antioxidants in AD. Moreover, this review may help researchers to develop effectively and potentially improved antioxidant therapeutic strategies for this disease as it also deals with the clinical trials in the stated field.
Collapse
|
18
|
S100B dysregulation during brain development affects synaptic SHANK protein networks via alteration of zinc homeostasis. Transl Psychiatry 2021; 11:562. [PMID: 34741005 PMCID: PMC8571423 DOI: 10.1038/s41398-021-01694-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 11/08/2022] Open
Abstract
Autism Spectrum Disorders (ASD) are caused by a combination of genetic predisposition and nongenetic factors. Among the nongenetic factors, maternal immune system activation and zinc deficiency have been proposed. Intriguingly, as a genetic factor, copy-number variations in S100B, a pro-inflammatory damage-associated molecular pattern (DAMP), have been associated with ASD, and increased serum S100B has been found in ASD. Interestingly, it has been shown that increased S100B levels affect zinc homeostasis in vitro. Thus, here, we investigated the influence of increased S100B levels in vitro and in vivo during pregnancy in mice regarding zinc availability, the zinc-sensitive SHANK protein networks associated with ASD, and behavioral outcomes. We observed that S100B affects the synaptic SHANK2 and SHANK3 levels in a zinc-dependent manner, especially early in neuronal development. Animals exposed to high S100B levels in utero similarly show reduced levels of free zinc and SHANK2 in the brain. On the behavioral level, these mice display hyperactivity, increased stereotypic and abnormal social behaviors, and cognitive impairment. Pro-inflammatory factors and zinc-signaling alterations converge on the synaptic level revealing a common pathomechanism that may mechanistically explain a large share of ASD cases.
Collapse
|
19
|
Volpina OM, Koroev DO, Serebryakova MV, Volkova TD, Kamynina AV, Bobkova NV. Proteolytic degradation patterns of the receptor for advanced glycation end products peptide fragments correlate with their neuroprotective activity in Alzheimer's disease models. Drug Dev Res 2021; 82:1217-1226. [PMID: 34060112 DOI: 10.1002/ddr.21836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 11/10/2022]
Abstract
The receptor for advanced glycation end products (RAGE) plays an essential role in Alzheimer's disease (AD). We previously demonstrated that a fragment (60-76) of RAGE improved the memory of olfactory bulbectomized (OBX) and Tg 5 × FAD mice - animal models of AD. The peptide analog (60-76) with protected N- and C-terminal groups was more active than the free peptide in Tg 5 × FAD mice. This study investigated proteolytic cleavage of the RAGE fragment (60-76) and its C- and N-terminally modified analog by blood serum using HPLC and mass spectrometry. The modified peptide was proteolyzed slower than the free peptide. Degrading the protected analog resulted in shortened fragments with memory-enhancing effects, whereas the free peptide yielded inactive fragments. After administering the different peptides to OBX mice, their performance in a spatial memory task revealed that the effective dose of the modified peptide was five times lower than that of the free peptide. HPLC and mass spectrometry analysis of the proteolytic products allowed us to clarify the differences in the neuroprotective activity conferred by administering these two peptides to AD animal models. The current study suggests that the modified RAGE fragment is more promising for the development of anti-AD therapy than its free analog.
Collapse
Affiliation(s)
- Olga M Volpina
- Department of Molecular Neurobiology, Laboratory of Synthetic Vaccines, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry (RAS), Moscow, Russia
| | - Dmitriy O Koroev
- Department of Molecular Neurobiology, Laboratory of Synthetic Vaccines, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry (RAS), Moscow, Russia
| | - Marina V Serebryakova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Tatyana D Volkova
- Department of Molecular Neurobiology, Laboratory of Synthetic Vaccines, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry (RAS), Moscow, Russia
| | - Anna V Kamynina
- Department of Molecular Neurobiology, Laboratory of Synthetic Vaccines, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry (RAS), Moscow, Russia.,Research Center for Molecular Mechanisms of Aging and Age Related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Natalia V Bobkova
- Laboratory of Cellular Mechanisms of Memory Pathology, Institute of Cell Biophysics (RAS), Pushchino, Russia
| |
Collapse
|
20
|
Kubis-Kubiak A, Wiatrak B, Piwowar A. The Impact of High Glucose or Insulin Exposure on S100B Protein Levels, Oxidative and Nitrosative Stress and DNA Damage in Neuron-Like Cells. Int J Mol Sci 2021; 22:ijms22115526. [PMID: 34073816 PMCID: PMC8197274 DOI: 10.3390/ijms22115526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/25/2022] Open
Abstract
Alzheimer’s disease (AD) is attracting considerable interest due to its increasing number of cases as a consequence of the aging of the global population. The mainstream concept of AD neuropathology based on pathological changes of amyloid β metabolism and the formation of neurofibrillary tangles is under criticism due to the failure of Aβ-targeting drug trials. Recent findings have shown that AD is a highly complex disease involving a broad range of clinical manifestations as well as cellular and biochemical disturbances. The past decade has seen a renewed importance of metabolic disturbances in disease-relevant early pathology with challenging areas in establishing the role of local micro-fluctuations in glucose concentrations and the impact of insulin on neuronal function. The role of the S100 protein family in this interplay remains unclear and is the aim of this research. Intracellularly the S100B protein has a protective effect on neurons against the toxic effects of glutamate and stimulates neurites outgrowth and neuronal survival. At high concentrations, it can induce apoptosis. The aim of our study was to extend current knowledge of the possible impact of hyper-glycemia and -insulinemia directly on neuronal S100B secretion and comparison to oxidative stress markers such as ROS, NO and DBSs levels. In this paper, we have shown that S100B secretion decreases in neurons cultured in a high-glucose or high-insulin medium, while levels in cell lysates are increased with statistical significance. Our findings demonstrate the strong toxic impact of energetic disturbances on neuronal metabolism and the potential neuroprotective role of S100B protein.
Collapse
Affiliation(s)
- Adriana Kubis-Kubiak
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
- Correspondence:
| | - Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland;
| | - Agnieszka Piwowar
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
| |
Collapse
|
21
|
Moysa A, Steczkiewicz K, Niedzialek D, Hammerschmid D, Zhukova L, Sobott F, Dadlez M. A model of full-length RAGE in complex with S100B. Structure 2021; 29:989-1002.e6. [PMID: 33887170 DOI: 10.1016/j.str.2021.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 02/03/2021] [Accepted: 04/02/2021] [Indexed: 01/10/2023]
Abstract
The receptor for advanced glycation end products (RAGE) is an immunoglobulin-type multiligand transmembrane protein expressed in numerous cell types, including the central nervous system cells. RAGE interaction with S100B, released during brain tissue damage, leads to RAGE upregulation and initialization of a spiral proinflammatory associated with different neural disorders. Here, we present the structural characterization of the hetero-oligomeric complex of the full-length RAGE with S100B, obtained by a combination of mass spectrometry-based methods and molecular modeling. We predict that RAGE functions as a tightly packed tetramer exposing a positively charged surface formed by V domains for S100B binding. Based on HDX results we demonstrate an allosteric coupling of the distal extracellular V domains and the transmembrane region, indicating a possible mechanism of signal transmission by RAGE across the membrane. Our model provides an insight into RAGE-ligand interactions, providing a basis for the rational design of the therapeutic modifiers of its activity.
Collapse
Affiliation(s)
- Alexander Moysa
- Institute of Biochemistry and Biophysics, PAS, Pawinskiego 5a, 02-109 Warsaw, Poland.
| | - Kamil Steczkiewicz
- Institute of Biochemistry and Biophysics, PAS, Pawinskiego 5a, 02-109 Warsaw, Poland.
| | - Dorota Niedzialek
- Institute of Biochemistry and Biophysics, PAS, Pawinskiego 5a, 02-109 Warsaw, Poland
| | - Dietmar Hammerschmid
- Department of Chemistry, King's College London, 7 Trinity Street, SE1 1DB London, UK; Department of Chemistry, Biomolecular & Analytical Mass Spectrometry Group, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Lilia Zhukova
- Institute of Biochemistry and Biophysics, PAS, Pawinskiego 5a, 02-109 Warsaw, Poland
| | - Frank Sobott
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Woodhouse Lane, LS2 9JT Leeds, UK; Department of Chemistry, Biomolecular & Analytical Mass Spectrometry Group, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Michal Dadlez
- Institute of Biochemistry and Biophysics, PAS, Pawinskiego 5a, 02-109 Warsaw, Poland
| |
Collapse
|
22
|
Langeh U, Singh S. Targeting S100B Protein as a Surrogate Biomarker and its Role in Various Neurological Disorders. Curr Neuropharmacol 2021; 19:265-277. [PMID: 32727332 PMCID: PMC8033985 DOI: 10.2174/1570159x18666200729100427] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/09/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023] Open
Abstract
Neurological disorders (ND) are the central nervous system (CNS) related complications originated by enhanced oxidative stress, mitochondrial failure and overexpression of proteins like S100B. S100B is a helix-loop-helix protein with the calcium-binding domain associated with various neurological disorders through activation of the MAPK pathway, increased NF-kB expression resulting in cell survival, proliferation and gene up-regulation. S100B protein plays a crucial role in Alzheimer's disease, Parkinson's disease, multiple sclerosis, Schizophrenia and epilepsy because the high expression of this protein directly targets astrocytes and promotes neuroinflammation. Under stressful conditions, S100B produces toxic effects mediated through receptor for advanced glycation end products (AGE) binding. S100B also mediates neuroprotection, minimizes microgliosis and reduces the expression of tumor necrosis factor (TNF-alpha) but that are concentration- dependent mechanisms. Increased level of S100B is useful for assessing the release of inflammatory markers, nitric oxide and excitotoxicity dependent neuronal loss. The present review summarizes the role of S100B in various neurological disorders and potential therapeutic measures to reduce the prevalence of neurological disorders.
Collapse
Affiliation(s)
- Urvashi Langeh
- Department of Neuropharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Department of Neuropharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
23
|
Sinyor B, Mineo J, Ochner C. Alzheimer's Disease, Inflammation, and the Role of Antioxidants. J Alzheimers Dis Rep 2020; 4:175-183. [PMID: 32715278 PMCID: PMC7369138 DOI: 10.3233/adr-200171] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2020] [Indexed: 12/24/2022] Open
Abstract
The World Health Organization refers to Alzheimer's disease (AD) as a global health priority. As the average age of the world's population is increasing, so too is the rate of AD. There are an estimated 47 million people globally who have been diagnosed with AD dementia, and researchers have yet to figure out the root cause. All misfolded aggregate proteins that are involved in neurodegenerative disorders (amyloid-β, Huntington's tau, α-synuclein) induce oxidative stress. It is that oxidative stress that leads to inflammation and, in conjunction with amyloid protein and tau hyperphosphorylation, progresses to and exacerbates AD. The consumption of antioxidants and nutrients, specifically vitamin E, caffeine, and turmeric, may slow the progression of AD and can be found in a wide variety of dietary foods. This review explores the role of inflammation on AD, the antioxidants that can potentially combat it, and future directions of how the treatment of the disease can be better understood.
Collapse
Affiliation(s)
- Benjamin Sinyor
- HCA Healthcare, Nashville, TN, USA
- Aventura Hospital and Medical Center, Aventura, FL, USA
| | - Jocelyn Mineo
- HCA Healthcare, Nashville, TN, USA
- Aventura Hospital and Medical Center, Aventura, FL, USA
| | - Christopher Ochner
- HCA Healthcare, Nashville, TN, USA
- Aventura Hospital and Medical Center, Aventura, FL, USA
| |
Collapse
|
24
|
Koroev DO, Volpina OM, Volkova TD, Kamynina AV, Samokhin AN, Filatova MP, Bobkova NV. A Synthetic Fragment of the Receptor for Glycation End Products and Its Analogue Improve Memory in Transgenic Alzheimer’s Disease Mouse Model. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2019. [DOI: 10.1134/s1068162019050054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
25
|
Hagmeyer S, Romão MA, Cristóvão JS, Vilella A, Zoli M, Gomes CM, Grabrucker AM. Distribution and Relative Abundance of S100 Proteins in the Brain of the APP23 Alzheimer's Disease Model Mice. Front Neurosci 2019; 13:640. [PMID: 31281238 PMCID: PMC6596341 DOI: 10.3389/fnins.2019.00640] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence links proteins of the S100 family to the pathogenesis of Alzheimer's disease (AD). S100 proteins are EF-hand calcium-binding proteins with intra- and extracellular functions related to regulation of proliferation, differentiation, apoptosis, and trace metal homeostasis, and are important modulators of inflammatory responses. For example, S100A6, S100A8, and S100B expression levels were found increased in inflammatory diseases, but also neurodegenerative disorders, and S100A8/A9 complexes may provide a mechanistic link between amyloid-beta (Aβ) plaque formation and neuroinflammation. On the other hand, S100B, a proinflammatory protein that is chronically up-regulated in AD and whose elevation precedes plaque formation, was recently shown to suppress Aβ aggregation. Here, we report expression of S100A6 and S100B in astrocytes and less so in neurons, and low level of expression of S100A8 in both neurons and glial cells in vitro. In vivo, S100A8 expression is almost absent in the brain of aged wildtype mice, while S100A6 and S100B are expressed in all brain regions and most prominently in the cortex and cerebellum. S100B seems to be enriched in Purkinje cells of the cerebellum. In contrast, in the brain of APP23 mice, a mouse model for Alzheimer's disease, S100B, S100A6, and S100A8 show co-localization with Aβ plaques, compatible with astrocyte activation, and the expression level of S100A8 is increased in neural cells. While S100A6 and S100B are enriched in the periphery of plaques where less fibrillar Aβ is found, S100A8 is more intense within the center of the inclusion. In vitro assays show that, similarly to S100B, S100A6, and S100A8 also delay Aβ aggregation suggesting a regulatory action over protein aggregation. We posit that elevated expression levels and overlapping spatial distribution of brain S100 proteins and plaques translates functional relationships between these inflammatory mediators and AD pathophysiology processes that uncover important molecular mechanisms linking the aggregation and neuroinflammation cascades.
Collapse
Affiliation(s)
- Simone Hagmeyer
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, Limerick, Ireland
- WG Molecular Analysis of Synaptopathies, Department of Neurology, Neurocenter of Ulm University, Ulm, Germany
| | - Mariana A. Romão
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Departamento de Química e Bioquímica, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S. Cristóvão
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Departamento de Química e Bioquímica, Universidade de Lisboa, Lisbon, Portugal
| | - Antonietta Vilella
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - Michele Zoli
- Department of Biomedical, Metabolic and Neural Sciences, Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - Cláudio M. Gomes
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Departamento de Química e Bioquímica, Universidade de Lisboa, Lisbon, Portugal
| | - Andreas M. Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
26
|
Abstract
During the past decades, life expectancy of subjects with Down syndrome (DS) has greatly improved, but age-specific mortality rates are still important and DS subjects are characterized by an acceleration of the ageing process, which affects particularly the immune and central nervous systems. In this chapter, we will first review the characteristics of the ageing phenomenon in brain and in immune system in DS and we will then discuss the biological hallmarks of ageing in this specific population. Finally, we will also consider in detail the knowledge on epigenetics in DS, particularly DNA methylation.
Collapse
|
27
|
Volpina OM, Samokhin AN, Koroev DO, Nesterova IV, Volkova TD, Medvinskaya NI, Nekrasov PV, Tatarnikova OG, Kamynina AV, Balasanyants SM, Voronina TA, Kulikov AM, Bobkova NV. Synthetic Fragment of Receptor for Advanced Glycation End Products Prevents Memory Loss and Protects Brain Neurons in Olfactory Bulbectomized Mice. J Alzheimers Dis 2019; 61:1061-1076. [PMID: 29332040 DOI: 10.3233/jad-170483] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Activation of receptor for advanced glycation end products (RAGE) plays an essential role in the development of Alzheimer's disease (AD). It is known that the soluble isoform of the receptor binds to ligands and prevents negative effects of the receptor activation. We proposed that peptide fragments from RAGE prevent negative effects of the receptor activation during AD neurodegeneration. We have synthesized peptide fragments from surface-exposed regions of RAGE. Peptides were intranasally administrated into olfactory bulbectomized (OBX) mice, which developed some characteristics similar to AD neurodegeneration. We have found that only insertion of fragment (60-76) prevents the memory of OBX mice. Immunization of OBX mice with peptides showed that again only (60-76) peptide protected the memory of animals. Both intranasal insertion and immunization decreased the amyloid-β (Aβ) level in the brain. Activity of shortened fragments of (60-76) peptide was tested and showed only the (60-70) peptide is responsible for manifestation of activity. Intranasal administration of (60-76) peptide shows most protective effect on morpho-functional characteristics of neurons in the cortex and hippocampal areas. Using Flu-(60-76) peptide, we revealed its penetration in the brain of OBX mice as well as colocalization of Flu-labeled peptide with Aβ in the brain regions in transgenic mice. Flu-(60-76) peptide complex with trimer of Aβ was detected by SDS-PAGE. These data indicate that Aβ can be one of the molecular target of (60-70) peptide. These findings provide a new peptide molecule for design of anti-AD drug and for investigation of RAGE activation ways in progression of AD neurodegeneration.
Collapse
Affiliation(s)
- Olga M Volpina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexandr N Samokhin
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Dmitriy O Koroev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Inna V Nesterova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Tatyana D Volkova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Pavel V Nekrasov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Olga G Tatarnikova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Anna V Kamynina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Samson M Balasanyants
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Alexey M Kulikov
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Natalia V Bobkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|
28
|
Wang Y, Tadayon R, Shaw GS. Monitoring Interactions Between S100B and the Dopamine D2 Receptor Using NMR Spectroscopy. Methods Mol Biol 2019; 1929:311-324. [PMID: 30710282 DOI: 10.1007/978-1-4939-9030-6_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
S100B is a dimeric EF-hand protein that undergoes a calcium-induced conformational change and interacts with a wide range of proteins to modulate their functions. The dopamine D2 receptor is one potential S100B binding partner that may play a key role in neurological processing. In this chapter, we describe the use of NMR spectroscopy to examine the interaction between calcium-bound S100B and the third intracellular loop (IC3) from the dopamine D2 receptor. We provide details that allow the strength of the interaction (K d) between the two proteins to be determined and the IC3 site of interaction on the structure of S100B to be identified. Both these characteristics can be identified from a single series of nondestructive experiments.
Collapse
Affiliation(s)
- Yuning Wang
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Roya Tadayon
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada
| | - Gary S Shaw
- Department of Biochemistry, The University of Western Ontario, London, ON, Canada.
| |
Collapse
|
29
|
Kamynina AV, Esteras N, Koroev DO, Bobkova NV, Balasanyants SM, Simonyan RA, Avetisyan AV, Abramov AY, Volpina OM. Synthetic Fragments of Receptor for Advanced Glycation End Products Bind Beta-Amyloid 1-40 and Protect Primary Brain Cells From Beta-Amyloid Toxicity. Front Neurosci 2018; 12:681. [PMID: 30319347 PMCID: PMC6170785 DOI: 10.3389/fnins.2018.00681] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/10/2018] [Indexed: 11/18/2022] Open
Abstract
Receptor for advanced glycation end products (RAGE) is involved in the pathogenesis of Alzheimer's disease. We have previously revealed that RAGE fragment sequence (60-76) and its shortened analogs sequence (60-70) and (60-65) under intranasal insertion were able to restore memory and improve morphological and biochemical state of neurons in the brain of bulbectomized mice developing major AD features. In the current study, we have investigated the ability of RAGE peptide (60-76) and five shortened analogs to bind beta-amyloid (Aβ) 1-40 in an fluorescent titration test and show that all the RAGE fragments apart from one [sequence (65-76)] were able to bind Aβ in vitro. Moreover, we show that all RAGE fragments apart from the shortest one (60-62), were able to protect neuronal primary cultures from amyloid toxicity, by preventing the caspase 3 activation induced by Aβ 1-42. We have compared the data obtained in the present research with the previously published data in the animal model of AD, and offer a probable mechanism of neuroprotection of the RAGE peptide.
Collapse
Affiliation(s)
- Anna V. Kamynina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Noemi Esteras
- Department of Clinical and Movement Neurosciences, Institute of Neurology, University College London, London, United Kingdom
| | - Dmitriy O. Koroev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Natalia V. Bobkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Samson M. Balasanyants
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ruben A. Simonyan
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - Armine V. Avetisyan
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - Andrey Y. Abramov
- Department of Clinical and Movement Neurosciences, Institute of Neurology, University College London, London, United Kingdom
| | - Olga M. Volpina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
30
|
Chen M, Lee HK, Moo L, Hanlon E, Stein T, Xia W. Common proteomic profiles of induced pluripotent stem cell-derived three-dimensional neurons and brain tissue from Alzheimer patients. J Proteomics 2018; 182:21-33. [PMID: 29709615 DOI: 10.1016/j.jprot.2018.04.032] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/03/2018] [Accepted: 04/24/2018] [Indexed: 01/21/2023]
Abstract
We established a unique platform for proteomic analysis of cultured three-dimensional (3D) neurons and brain tissue from Alzheimer's disease (AD) patients. We collected peripheral blood mononuclear cells (PBMC), converted PBMC to induced pluripotent stem cell (iPSC) lines, and differentiated the iPSC into human 3D neuro-spheroids. The postmortem brain tissue from the superior frontal cortex, inferior frontal cortex and cerebellum area of the AD patients was compared to the same regions from the control subjects. Proteomic analysis of 3D neuro-spheroids derived from AD subjects revealed the alteration of a number of proteins involved in axon growth, mitochondrial function, and antioxidant defense. Similar analysis of post-mortem AD brain tissue revealed significant alteration in proteins involved in oxidative stress, neuro-inflammation, along with proteins related to axonal injury. These results clearly indicate that the dysfunction of 3D neurons from AD patients in our in vitro environment is comparable to the post-mortem AD brain tissue in vivo. In conclusion, our study revealed a number of candidate proteins that have important implications in AD pathogenesis and supports the notion that the iPSC-derived 3D neuronal system functions as a model to examine novel aspects of AD pathology. SIGNIFICANCE In this study, we present a unique platform for proteomic analysis of induced pluripotent stem cell-derived three dimensional (3D) neurons and compare the results to those from three regions of post-mortem brain tissue from Alzheimer's disease patients and normal control subjects. Our results show that the dysfunction of 3D neurons from AD patients in our in vitro environment is comparable to the post-mortem AD brain tissue in vivo. Our results revealed several candidate proteins that have important implications in AD pathogenesis.
Collapse
Affiliation(s)
- Mei Chen
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States; Department of Environmental Health, Harvard T H Chan School of Public Health, Boston, MA, United States
| | - Han-Kyu Lee
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States
| | - Lauren Moo
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States
| | - Eugene Hanlon
- Office of Research and Development, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States
| | - Thor Stein
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States; Department of Pathology, Boston University School of Medicine, Boston, MA, United States
| | - Weiming Xia
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States.
| |
Collapse
|
31
|
Ferguson SA, Panos JJ, Sloper D, Varma V. Neurodegenerative Markers are Increased in Postmortem BA21 Tissue from African Americans with Alzheimer's Disease. J Alzheimers Dis 2018; 59:57-66. [PMID: 28582866 DOI: 10.3233/jad-170204] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) presents with an earlier onset age and increased symptom severity in African Americans and Hispanics. OBJECTIVE Although the prevalence of plaques and tangles may not exhibit ethnicity-related differences, levels of neurodegenerative proteins have not been described. METHODS Here, levels of five proteins (i.e., S100B, sRAGE, GDNF, Aβ40, and Aβ42) and the Aβ42/Aβ40 ratio were measured in postmortem samples of the middle temporal gyrus (BA21) from age-matched African Americans and Caucasians with AD (n = 6/gender/ethnicity). RESULTS S100B levels were increased 17% in African Americans (p < 0.003) while sRAGE was mildly decreased (p < 0.09). Aβ42 levels were increased 121% in African Americans (p < 0.02), leading to a 493% increase in the Aβ42/Aβ40 ratio (p < 0.002). Analysis of GDNF levels did not indicate any significant effects. There were no significant effects of gender and no significant ethnicity with gender interactions on any analyte. Effect size calculations indicated "medium" to "very large" effects. CONCLUSION S100B is typically elevated in AD cases; however, the increased levels in African Americans here may be indicative of increased severity in specific populations. Increased Aβ42/Aβ40 ratios in the current study are compatible with increased disease severity and might indicate increased AD pathogenesis in African Americans. Overall, these results are compatible with a hypothesis of increased neuroinflammation in African Americans with AD.
Collapse
Affiliation(s)
- Sherry A Ferguson
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, USA
| | - John J Panos
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, USA
| | - Daniel Sloper
- Division of Systems Biology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, USA
| | - Vijayalakshmi Varma
- Division of Systems Biology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
32
|
Metabolic Derangements Contribute to Reduced sRAGE Isoforms in Subjects with Alzheimer's Disease. Mediators Inflamm 2018; 2018:2061376. [PMID: 29681765 PMCID: PMC5842684 DOI: 10.1155/2018/2061376] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/09/2018] [Indexed: 01/20/2023] Open
Abstract
Although there is evidence for metabolic dysfunction and chronic inflammation in Alzheimer's disease (AD), circulating levels of soluble receptor for advanced glycation end products (sRAGE) and the receptor for advanced glycation end products (RAGE) ligand S100B have not been characterized. sRAGE is an important mediator in disease as it can act as a ligand decoy for RAGE and attenuate downstream inflammatory signaling. Cognitively healthy elderly and AD participants with and without type 2 diabetes (n = 135) were stratified according to the clinical dementia rating (CDR; 0 = normal cognition (NC); ≥0.5 = AD). Total serum sRAGE, endogenous secretory RAGE (esRAGE), and S100B were assayed via ELISAs, and cleaved RAGE (cRAGE) and the cRAGE : esRAGE ratio were calculated. cRAGE : esRAGE was lower in AD compared to NC (p < 0.05). Metabolic substratifications were used to investigate the factors that influence sRAGE pathology in AD. Stratification by BMI classification, median fat mass, median HOMA-IR, median insulin, and median amylin were all metabolic or anthropometric factors which significantly interacted with sRAGE profiles within AD subjects. There were no significant differences in serum S100B between groups. These characterizations of sRAGE contribute evidence to the link between impaired metabolism and cognitive decline due to AD.
Collapse
|
33
|
Hagmeyer S, Cristóvão JS, Mulvihill JJE, Boeckers TM, Gomes CM, Grabrucker AM. Zinc Binding to S100B Affords Regulation of Trace Metal Homeostasis and Excitotoxicity in the Brain. Front Mol Neurosci 2018; 10:456. [PMID: 29386995 PMCID: PMC5776125 DOI: 10.3389/fnmol.2017.00456] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/26/2017] [Indexed: 12/16/2022] Open
Abstract
Neuronal metal ions such as zinc are essential for brain function. In particular synaptic processes are tightly related to metal and protein homeostasis, for example through extracellular metal-binding proteins. One such protein is neuronal S100B, a calcium and zinc binding damage-associated molecular pattern (DAMP), whose chronic upregulation is associated with aging, Alzheimer’s disease (AD), motor neuron disease and traumatic brain injury (TBI). Despite gained insights on the structure of S100B, it remains unclear how its calcium and zinc binding properties regulate its function on cellular level. Here we report a novel role of S100B in trace metal homeostasis, in particular the regulation of zinc levels in the brain. Our results show that S100B at increased extracellular levels is not toxic, persists at high levels, and is taken up into neurons, as shown by cell culture and biochemical analysis. Combining protein bioimaging and zinc quantitation, along with a zinc-binding impaired S100B variant, we conclude that S100B effectively scavenges zinc ions through specific binding, resulting in a redistribution of the intracellular zinc pool. Our results indicate that scavenging of zinc by increased levels of S100B affects calcium levels in vitro. Thereby S100B is able to mediate the cross talk between calcium and zinc homeostasis. Further, we investigated a possible new neuro-protective role of S100B in excitotoxicity via its effects on calcium and zinc homeostasis. Exposure of cells to zinc-S100B but not the zinc-binding impaired S100B results in an inhibition of excitotoxicity. We conclude that in addition to its known functions, S100B acts as sensor and regulator of elevated zinc levels in the brain and this metal-buffering activity is tied to a neuroprotective role.
Collapse
Affiliation(s)
- Simone Hagmeyer
- WG Molecular Analysis of Synaptopathies, Department of Neurology, Neurocenter of Ulm University, Ulm, Germany.,Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland
| | - Joana S Cristóvão
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - John J E Mulvihill
- Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Cláudio M Gomes
- Biosystems and Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Andreas M Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
34
|
Wang C, Shou Y, Pan J, Du Y, Liu C, Wang H. The relationship between cholesterol level and Alzheimer’s disease-associated APP proteolysis/Aβ metabolism. Nutr Neurosci 2018; 22:453-463. [DOI: 10.1080/1028415x.2017.1416942] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Chaoqun Wang
- School of Medicine, Hangzhou Normal University, Hangzhou, People’s Republic of China
| | - Yikai Shou
- School of Medicine, Hangzhou Normal University, Hangzhou, People’s Republic of China
| | - Jie Pan
- Department of Endocrinology and Metabolism, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, People’s Republic of China
| | - Yue Du
- School of Medicine, Zhejiang University, Hangzhou, People’s Republic of China
| | - Cuiqing Liu
- College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Huanhuan Wang
- School of Medicine, Hangzhou Normal University, Hangzhou, People’s Republic of China
| |
Collapse
|
35
|
Oh S, Son M, Choi J, Lee S, Byun K. sRAGE prolonged stem cell survival and suppressed RAGE-related inflammatory cell and T lymphocyte accumulations in an Alzheimer's disease model. Biochem Biophys Res Commun 2018; 495:807-813. [DOI: 10.1016/j.bbrc.2017.11.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 11/06/2017] [Indexed: 01/08/2023]
|
36
|
Hov KR, Bolstad N, Idland AV, Zetterberg H, Blennow K, Chaudhry FA, Frihagen F, Ræder J, Wyller TB, Watne LO. Cerebrospinal Fluid S100B and Alzheimer's Disease Biomarkers in Hip Fracture Patients with Delirium. Dement Geriatr Cogn Dis Extra 2017; 7:374-385. [PMID: 29282410 PMCID: PMC5731172 DOI: 10.1159/000481853] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/19/2017] [Indexed: 01/23/2023] Open
Abstract
Objectives This study aimed to investigate the relationship between cerebrospinal fluid (CSF) S100B astrocyte-derived protein and delirium and to perform stratified analyses according to clinical and CSF markers of dementia. Methods We performed a prospective cohort study in a university hospital setting. The participants were patients admitted for hip fracture (n = 98) or for elective surgery (n = 50). Delirium was assessed daily perioperatively in hip fracture patients using the Confusion Assessment Method. A consensus-based diagnosis of prefracture dementia was made using all available information. CSF was drawn at the onset of spinal anesthesia. S100B and phosphorylated tau (P-tau) concentrations were measured using electrochemiluminescence immunoassay and enzyme-linked immunosorbent assays, respectively. Results In the hip fracture population (n = 98) there was no significant difference in CSF S100B concentrations between patients with ongoing preoperative (i.e., prevalent) delirium (n = 36, median [interquartile range] 1.11 μg/L [0.91–1.29]) and patients who never developed delirium (n = 46, 1.08 μg/L [0.92–1.28], p = 0.92). In patients without preoperative delirium, those who developed delirium postoperatively (i.e., incident delirium) (n = 16, 1.38 μg/L [1.08–1.62]) had higher concentrations of S100B than the 46 who never did (p = 0.013). This difference was confined to patients with pathological concentrations of P-tau (≥60 ng/L, n = 38). We also found that P-tau and S100B were correlated in CSF in the elective surgery patients. Conclusions CSF S100B was elevated in patients with incident delirium who also had pathological levels of the Alzheimer disease biomarker P-tau, suggesting vulnerability caused by a preexisting process of astrocytic activation and tau pathology.
Collapse
Affiliation(s)
- Karen Roksund Hov
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nils Bolstad
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Ane-Victoria Idland
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, United Kingdom
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Farrukh A Chaudhry
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Frede Frihagen
- Division of Orthopedic Surgery, Oslo University Hospital, Oslo, Norway
| | - Johan Ræder
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Anesthesiology, Oslo University Hospital, Oslo, Norway
| | - Torgeir Bruun Wyller
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Leiv Otto Watne
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
37
|
Son M, Oh S, Park H, Ahn H, Choi J, Kim H, Lee HS, Lee S, Park HJ, Kim SU, Lee B, Byun K. Protection against RAGE-mediated neuronal cell death by sRAGE-secreting human mesenchymal stem cells in 5xFAD transgenic mouse model. Brain Behav Immun 2017; 66:347-358. [PMID: 28760504 DOI: 10.1016/j.bbi.2017.07.158] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 07/26/2017] [Accepted: 07/27/2017] [Indexed: 10/19/2022] Open
Abstract
Alzheimer's disease (AD), which is the most commonly encountered neurodegenerative disease, causes synaptic dysfunction and neuronal loss due to various pathological processes that include tau abnormality and amyloid beta (Aβ) accumulation. Aβ stimulates the secretion and the synthesis of Receptor for Advanced Glycation End products (RAGE) ligand by activating microglial cells, and has been reported to cause neuronal cell death in Aβ1-42 treated rats and in mice with neurotoxin-induced Parkinson's disease. The soluble form of RAGE (sRAGE) is known to reduce inflammation, and to decrease microglial cell activation and Aβ deposition, and thus, it protects from neuronal cell death in AD. However, sRAGE protein has too a short half-life for therapeutic purposes. We developed sRAGE-secreting umbilical cord derived mesenchymal stem cells (sRAGE-MSCs) to enhance the inhibitory effects of sRAGE on Aβ deposition and to reduce the secretion and synthesis of RAGE ligands in 5xFAD mice. In addition, these cells improved the viability of injected MSCs, and enhanced the protective effects of sRAGE by inhibiting the binding of RAGE and RAGE ligands in 5xFAD mice. These findings suggest sRAGE protein from sRAGE-MSCs has better protection against neuronal cell death than sRAGE protein or single MSC treatment by inhibiting the RAGE cell death cascade and RAGE-induce inflammation.
Collapse
Affiliation(s)
- Myeongjoo Son
- Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea; Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Hyunjin Park
- Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea; Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Hyosang Ahn
- Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea; Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Junwon Choi
- Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea; Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Hyungho Kim
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Hye Sun Lee
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Sojung Lee
- Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea; Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Hye-Jeong Park
- Department of Biochemistry and Cell Biology, Kyungpook National University, Daegu, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung U Kim
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Bonghee Lee
- Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea; Center for Regenerative Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.
| | - Kyunghee Byun
- Department of Anatomy and Cell Biology, Gachon University Graduate School of Medicine, Incheon, Republic of Korea; Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.
| |
Collapse
|
38
|
D'Cunha NM, McKune AJ, Panagiotakos DB, Georgousopoulou EN, Thomas J, Mellor DD, Naumovski N. Evaluation of dietary and lifestyle changes as modifiers of S100β levels in Alzheimer's disease. Nutr Neurosci 2017; 22:1-18. [PMID: 28696163 DOI: 10.1080/1028415x.2017.1349032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There is a significant body of research undertaken in order to elucidate the mechanisms underlying the pathology of Alzheimer's disease (AD), as well as to discover early detection biomarkers and potential therapeutic strategies. One such proposed biomarker is the calcium binding protein S100β, which, depending on its local concentration, is known to exhibit both neurotrophic and neuroinflammatory properties in the central nervous system. At present, relatively little is known regarding the effect of chronic S100β disruption in AD. Dietary intake has been identified as a modifiable risk factor for AD. Preliminary in vitro and animal studies have demonstrated an association between S100β expression and dietary intake which links to AD pathophysiology. This review describes the association of S100β to fatty acids, ketone bodies, insulin, and botanicals as well as the potential impact of physical activity as a lifestyle factor. We also discuss the prospective implications of these findings, including support of the use of a Mediterranean dietary pattern and/or the ketogenic diet as an approach to modify AD risk.
Collapse
Affiliation(s)
- Nathan M D'Cunha
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| | - Andrew J McKune
- b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia.,c University of Canberra, Research Institute for Sport and Exercise , University of Canberra , Bruce , Canberra ACT 2601 , Australia.,d Discipline of Biokinetics, Exercise and Leisure Sciences, School of Health Sciences , University of KwaZulu-Natal , Durban 4041 , South Africa
| | - Demosthenes B Panagiotakos
- e Department of Nutrition-Dietetics, School of Health and Education , Harokopio University , Athens 176 71 , Greece
| | - Ekavi N Georgousopoulou
- b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia.,e Department of Nutrition-Dietetics, School of Health and Education , Harokopio University , Athens 176 71 , Greece
| | - Jackson Thomas
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| | - Duane D Mellor
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| | - Nenad Naumovski
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| |
Collapse
|
39
|
Liu L, Yu J, Li L, Zhang B, Liu L, Wu CH, Jong A, Mao DA, Huang SH. Alpha7 nicotinic acetylcholine receptor is required for amyloid pathology in brain endothelial cells induced by Glycoprotein 120, methamphetamine and nicotine. Sci Rep 2017; 7:40467. [PMID: 28074940 PMCID: PMC5225415 DOI: 10.1038/srep40467] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/06/2016] [Indexed: 01/12/2023] Open
Abstract
One of the most challenging issues in HIV-associated neurocognitive disorders (HAND) caused by HIV-1 virotoxins and drug abuse is the lack of understanding the underlying mechanisms that are commonly associated with disorders of the blood-brain barrier (BBB), which mainly consists of brain microvascular endothelial cells (BMEC). Here, we hypothesized that Glycoprotein 120 (gp120), methamphetamine (METH) and nicotine (NT) can enhance amyloid-beta (Aβ) accumulation in BMEC through Alpha7 nicotinic acetylcholine receptor (α7 nAChR). Both in vitro (human BMEC) (HBMEC) and in vivo (mice) models of BBB were used to dissect the role of α7 nAChR in up-regulation of Aβ induced by gp120, METH and NT. Aβ release from and transport across HBMEC were significantly increased by these factors. Methyllycaconitine (MLA), an antagonist of α7 nAChR, could efficiently block these pathogenic effects. Furthermore, our animal data showed that these factors could significantly increase the levels of Aβ, Tau and Ubiquitin C-Terminal Hydrolase L1 (UCHL1) in mouse cerebrospinal fluid (CSF) and Aβ in the mouse brains. These pathogenicities were significantly reduced by MLA, suggesting that α7 nAChR may play an important role in neuropathology caused by gp120, METH and NT, which are the major pathogenic factors contributing to the pathogenesis of HAND.
Collapse
Affiliation(s)
- Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.,Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA90027, USA
| | - Jingyi Yu
- Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA90027, USA.,School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Li Li
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, 510515, China.,Department of Clinical Laboratory, Kunming Children's Hospital, Kunming Medical University, Kunming, Yunnan 650034, China
| | - Bao Zhang
- Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA90027, USA.,School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chun-Hua Wu
- Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA90027, USA
| | - Ambrose Jong
- Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA90027, USA
| | - Ding-An Mao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Sheng-He Huang
- Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA90027, USA.,School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
40
|
Shafi O. Inverse relationship between Alzheimer's disease and cancer, and other factors contributing to Alzheimer's disease: a systematic review. BMC Neurol 2016; 16:236. [PMID: 27875990 PMCID: PMC5120447 DOI: 10.1186/s12883-016-0765-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 11/15/2016] [Indexed: 12/18/2022] Open
Abstract
Background The AD etiology is yet not properly known. Interactions among environmental factors, multiple susceptibility genes and aging, contribute to AD. This study investigates the factors that play role in causing AD and how changes in cellular pathways contribute to AD. Methods PUBMED database, MEDLINE database and Google Scholar were searched with no date restrictions for published articles involving cellular pathways with roles in cancers, cell survival, growth, proliferation, development, aging, and also contributing to Alzheimer’s disease. This research explores inverse relationship between AD and cancer, also investigates other factors behind AD using several already published research literature to find the etiology of AD. Results Cancer and Alzheimer’s disease have inverse relationship in many aspects such as P53, estrogen, neurotrophins and growth factors, growth and proliferation, cAMP, EGFR, Bcl-2, apoptosis pathways, IGF-1, HSV, TDP-43, APOE variants, notch signals and presenilins, NCAM, TNF alpha, PI3K/AKT/MTOR pathway, telomerase, ROS, ACE levels. AD occurs when brain neurons have weakened growth, cell survival responses, maintenance mechanisms, weakened anti-stress responses such as Vimentin, Carbonic anhydrases, HSPs, SAPK. In cancer, these responses are upregulated and maintained. Evolutionarily conserved responses and maintenance mechanisms such as FOXO are impaired in AD. Countermeasures or compensatory mechanisms by AD affected neurons such as Tau, Beta Amyloid, S100, are last attempts for survival which may be protective for certain time, or can speed up AD in Alzheimer’s microenvironment via C-ABL activation, GSK3, neuro-inflammation. Conclusions Alzheimer’s disease and Cancer have inverse relationship; many factors that are upregulated in any cancer to sustain growth and survival are downregulated in Alzheimer’s disease contributing to neuro-degeneration. When aged neurons or genetically susceptible neurons have weakened growth, cell survival and anti-stress responses, age related gene expression changes, altered regulation of cell death and maintenance mechanisms, they contribute to Alzheimer’s disease. Countermeasures by AD neurons such as Beta Amyloid Plaques, NFTs, S100, are last attempts for survival and this provides neuroprotection for certain time and ultimately may become pathological and speed up AD. This study may contribute in developing new potential diagnostic tests, interventions and treatments. Electronic supplementary material The online version of this article (doi:10.1186/s12883-016-0765-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College, Dow University of Health Sciences, Karachi, Pakistan.
| |
Collapse
|
41
|
Créau N, Cabet E, Daubigney F, Souchet B, Bennaï S, Delabar J. Specific age-related molecular alterations in the cerebellum of Down syndrome mouse models. Brain Res 2016; 1646:342-353. [PMID: 27297494 DOI: 10.1016/j.brainres.2016.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/07/2016] [Accepted: 06/02/2016] [Indexed: 12/27/2022]
Abstract
Down syndrome, or trisomy 21, has been modeled with various trisomic and transgenic mice to help understand the consequences of an altered gene dosage in brain development and function. Though Down syndrome has been associated with premature aging, little is known about the molecular and cellular alterations that target brain function. To help identify alterations at specific ages, we analyzed the cerebellum of Ts1Cje mice, trisomic for 77 HSA21 orthologs, at three ages-young (4 months), middle-age (12 months), and old (17 months)-compared to age-matched controls. Quantification of neuronal and glial markers (n=11) revealed increases in GFAP, with an age effect, and S100B, with age and genotype effects. The genotype effect on S100B with age was unexpected as Ts1Cje has only two copies of the S100b gene. Interestingly, the different increase in GFAP observed between Ts1Cje (trisomic segment includes Pcp4 gene) and controls was magnified in TgPCP4 mice (1 extra copy of the human PCP4 gene) at the same age. S100B increase was not found in the TgPCP4 confirming a difference of regulation with aging for GFAP and S100B and excluding the calcium signaling regulator, Pcp4, as a potential candidate for increase of S100B in the Ts1Cje. To understand these differences, comparison of GFAP and S100B immunostainings at young and middle-age were performed. Immunohistochemical detection of differences in GFAP and S100B localization with aging implicate S100B+ oligodendrocytes as a new phenotypic target in this specific aging process.
Collapse
Affiliation(s)
- Nicole Créau
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, UMR8251, CNRS, Paris, France.
| | - Eva Cabet
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, UMR8251, CNRS, Paris, France
| | - Fabrice Daubigney
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, UMR8251, CNRS, Paris, France
| | - Benoit Souchet
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, UMR8251, CNRS, Paris, France
| | - Soumia Bennaï
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, UMR8251, CNRS, Paris, France
| | - Jean Delabar
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, UMR8251, CNRS, Paris, France
| |
Collapse
|
42
|
Festoff BW, Sajja RK, van Dreden P, Cucullo L. HMGB1 and thrombin mediate the blood-brain barrier dysfunction acting as biomarkers of neuroinflammation and progression to neurodegeneration in Alzheimer's disease. J Neuroinflammation 2016; 13:194. [PMID: 27553758 PMCID: PMC4995775 DOI: 10.1186/s12974-016-0670-z] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/17/2016] [Indexed: 01/11/2023] Open
Abstract
Background The blood-brain barrier (BBB) dysfunction represents an early feature of Alzheimer’s disease (AD) that precedes the hallmarks of amyloid beta (amyloid β) plaque deposition and neuronal neurofibrillary tangle (NFT) formation. A damaged BBB correlates directly with neuroinflammation involving microglial activation and reactive astrogliosis, which is associated with increased expression and/or release of high-mobility group box protein 1 (HMGB1) and thrombin. However, the link between the presence of these molecules, BBB damage, and progression to neurodegeneration in AD is still elusive. Therefore, we aimed to profile and validate non-invasive clinical biomarkers of BBB dysfunction and neuroinflammation to assess the progression to neurodegeneration in mild cognitive impairment (MCI) and AD patients. Methods We determined the serum levels of various proinflammatory damage-associated molecules in aged control subjects and patients with MCI or AD using validated ELISA kits. We then assessed the specific and direct effects of such molecules on BBB integrity in vitro using human primary brain microvascular endothelial cells or a cell line. Results We observed a significant increase in serum HMGB1 and soluble receptor for advanced glycation end products (sRAGE) that correlated well with amyloid beta levels in AD patients (vs. control subjects). Interestingly, serum HMGB1 levels were significantly elevated in MCI patients compared to controls or AD patients. In addition, as a marker of BBB damage, soluble thrombomodulin (sTM) antigen, and activity were significantly (and distinctly) increased in MCI and AD patients. Direct in vitro BBB integrity assessment further revealed a significant and concentration-dependent increase in paracellular permeability to dextrans by HMGB1 or α-thrombin, possibly through disruption of zona occludins-1 bands. Pre-treatment with anti-HMGB1 monoclonal antibody blocked HMGB1 effects and leaving BBB integrity intact. Conclusions Our current studies indicate that thrombin and HMGB1 are causal proximate proinflammatory mediators of BBB dysfunction, while sTM levels may indicate BBB endothelial damage; HMGB1 and sRAGE might serve as clinical biomarkers for progression and/or therapeutic efficacy along the AD spectrum.
Collapse
Affiliation(s)
- Barry W Festoff
- pHLOGISTIX LLC, 4220 Shawnee Mission Parkway, Fairway, KS, 66205, USA.,Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Ravi K Sajja
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Patrick van Dreden
- Clinical Research Department, R&D, Diagnostica Stago, Gennevilliers, France
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA.
| |
Collapse
|
43
|
ElAli A, Rivest S. Microglia in Alzheimer's disease: A multifaceted relationship. Brain Behav Immun 2016; 55:138-150. [PMID: 26254232 DOI: 10.1016/j.bbi.2015.07.021] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting elderly people worldwide, which is mainly characterized by cerebral amyloid-beta (Aβ) plaque deposition and neurofibrillary tangle formation. The interest in microglia arose from the overwhelming experimental evidence that outlined a key role of neuroinflammation in AD pathology. Microglia constitute the powerhouse of the innate immune system in the brain. It is now widely accepted that microglia are myeloid-derived cells that infiltrate the developing brain at the early embryonic stages, and acquire a highly ramified phenotype postnatally. Microglia use these dynamic ramifications as sentinels to sense and detect any occurring alteration in brain homeostasis. Once a danger signal is detected, microglia get activated by acquiring a less ramified phenotype, and mount adequate responses that range from phagocyting cell debris to secreting inflammatory and trophic factors. Earlier reports have demonstrated, unequivocally, that microglia surround Aβ plaques and internalize Aβ microaggregates. However, the implication of these observations in AD pathology, and consequently treatment, is still a matter of debate. Nonetheless, targeting the activity of these cells constituted a convergent point in this debate. Unfortunately, the conflicting experimental findings obtained following the modulation of microglial activity in AD, further fueled the debate. This review aims at providing an overview regarding what we know about the implication of microglia in AD pathology, and treatment. The emerging role of monocytes is also discussed.
Collapse
Affiliation(s)
- Ayman ElAli
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Canada.
| |
Collapse
|
44
|
Kurzątkowska K, Jankowska A, Wysłouch-Cieszyńska A, Zhukova L, Puchalska M, Dehaen W, Radecka H, Radecki J. Voltammetric detection of the S100B protein using His-tagged RAGE domain immobilized onto a gold electrode modified with a dipyrromethene–Cu(II) complex and different diluents. J Electroanal Chem (Lausanne) 2016. [DOI: 10.1016/j.jelechem.2016.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
45
|
GE YALI, LI XIAOBO, GAO JU, ZHANG XICHENG, FANG XIANGZHI, ZHOU LUOJING, JI WEI, LIN SHUNYAN. Beneficial effects of intravenous dexmedetomidine on cognitive function and cerebral injury following a carotid endarterectomy. Exp Ther Med 2016; 11:1128-1134. [PMID: 26998048 PMCID: PMC4774506 DOI: 10.3892/etm.2016.2978] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 11/25/2015] [Indexed: 12/27/2022] Open
Abstract
The present study aimed to investigate the effects of dexmedetomidine (DEX) on cognition following a carotid endarterectomy (CEA). In addition, the neuroprotective effects of DEX against ischemia-reperfusion injury during CEA were analyzed. Patients due to undergo elective CEA under general anesthesia were randomly assigned to either the DEX-treated group (group D; n=25) or the control group (group C; n=25). Patients in group D were treated with 0.3 µg/kg DEX pre-CEA, followed by 0.3 µg/kg/h DEX intraoperatively up to 30 min prior to the completion of surgery, and the patients in group C received an equal volume of normal saline. Cognitive function was assessed prior to CEA (T0), and at 24, 48, and 72 h, 7 days and 1 month post-surgery (T1-5, respectively), using the Mini-Mental State Examination (MMSE). Blood samples were drawn from the ipsilateral jugular bulb of all patients at 20 min prior to anesthesia (t0), and at 10 min following tracheal intubation, 15 min following clamping and unclamping of the carotid artery, and at 6 and 24 h postoperatively (t1-5, respectively). The protein expression levels of markers of cerebral ischemia and injury, namely S100 calcium-binding protein B (S100B) and neuron-specific enolase (NSE), and the concentration of the oxidative stress marker malondialdehyde (MDA), were analyzed. Patients in group D exhibited elevated MMSE scores at T2 and T3 post-CEA, as compared with group C. Furthermore, the protein expression level of S100B and the concentration of MDA in the jugular bulb of group D patients were markedly decreased compared with those in group C at t3-5 and t3, respectively. The results of the present study suggested that DEX was able to enhance the recovery of cognition following CEA, and this was associated with decreased cerebral damage and antioxidative effects.
Collapse
Affiliation(s)
- YA-LI GE
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - XIAOBO LI
- Department of Neurology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - JU GAO
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - XICHENG ZHANG
- Department of Vascular Surgery, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - XIANGZHI FANG
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - LUOJING ZHOU
- Department of Scientific Research, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - WEI JI
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - SHUNYAN LIN
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
46
|
Insulin Stimulates S100B Secretion and These Proteins Antagonistically Modulate Brain Glucose Metabolism. Neurochem Res 2016; 41:1420-9. [PMID: 26875731 DOI: 10.1007/s11064-016-1851-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 12/24/2022]
Abstract
Brain metabolism is highly dependent on glucose, which is derived from the blood circulation and metabolized by the astrocytes and other neural cells via several pathways. Glucose uptake in the brain does not involve insulin-dependent glucose transporters; however, this hormone affects the glucose influx to the brain. Changes in cerebrospinal fluid levels of S100B (an astrocyte-derived protein) have been associated with alterations in glucose metabolism; however, there is no evidence whether insulin modulates glucose metabolism and S100B secretion. Herein, we investigated the effect of S100B on glucose metabolism, measuring D-(3)H-glucose incorporation in two preparations, C6 glioma cells and acute hippocampal slices, and we also investigated the effect of insulin on S100B secretion. Our results showed that: (a) S100B at physiological levels decreases glucose uptake, through the multiligand receptor RAGE and mitogen-activated protein kinase/ERK signaling, and (b) insulin stimulated S100B secretion via PI3K signaling. Our findings indicate the existence of insulin-S100B modulation of glucose utilization in the brain tissue, and may improve our understanding of glucose metabolism in several conditions such as ketosis, streptozotocin-induced dementia and pharmacological exposure to antipsychotics, situations that lead to changes in insulin signaling and extracellular levels of S100B.
Collapse
|
47
|
Modeling the interaction between quinolinate and the receptor for advanced glycation end products (RAGE): relevance for early neuropathological processes. PLoS One 2015; 10:e0120221. [PMID: 25757085 PMCID: PMC4354912 DOI: 10.1371/journal.pone.0120221] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/20/2015] [Indexed: 01/13/2023] Open
Abstract
The receptor for advanced glycation end products (RAGE) is a pattern-recognition receptor involved in neurodegenerative and inflammatory disorders. RAGE induces cellular signaling upon binding to a variety of ligands. Evidence suggests that RAGE up-regulation is involved in quinolinate (QUIN)-induced toxicity. We investigated the QUIN-induced toxic events associated with early noxious responses, which might be linked to signaling cascades leading to cell death. The extent of early cellular damage caused by this receptor in the rat striatum was characterized by image processing methods. To document the direct interaction between QUIN and RAGE, we determined the binding constant (Kb) of RAGE (VC1 domain) with QUIN through a fluorescence assay. We modeled possible binding sites of QUIN to the VC1 domain for both rat and human RAGE. QUIN was found to bind at multiple sites to the VC1 dimer, each leading to particular mechanistic scenarios for the signaling evoked by QUIN binding, some of which directly alter RAGE oligomerization. This work contributes to the understanding of the phenomenon of RAGE-QUIN recognition, leading to the modulation of RAGE function.
Collapse
|
48
|
Afanador L, Roltsch EA, Holcomb L, Campbell KS, Keeling DA, Zhang Y, Zimmer DB. The Ca2+ sensor S100A1 modulates neuroinflammation, histopathology and Akt activity in the PSAPP Alzheimer's disease mouse model. Cell Calcium 2014; 56:68-80. [DOI: 10.1016/j.ceca.2014.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 11/25/2022]
|
49
|
Moser B, Janik S, Schiefer AI, Müllauer L, Bekos C, Scharrer A, Mildner M, Rényi-Vámos F, Klepetko W, Ankersmit HJ. Expression of RAGE and HMGB1 in thymic epithelial tumors, thymic hyperplasia and regular thymic morphology. PLoS One 2014; 9:e94118. [PMID: 24705787 PMCID: PMC3976415 DOI: 10.1371/journal.pone.0094118] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 03/13/2014] [Indexed: 02/07/2023] Open
Abstract
Recently, a role of the receptor for advanced glycation endproducts (RAGE) in myasthenia gravis was described. RAGE and its ligand high mobility group box 1 (HMGB1) play key roles in autoimmunity and cancer. To test whether these molecules are involved in patients with thymic abnormalities we applied immunohistochemical analysis in 33 cases of thymic epithelial tumors, comprising 27 thymomas and 6 thymic carcinomas, and 21 nonneoplastic thymuses. Both molecules were detected in neoplastic epithelial cells: RAGE staining was most intense in WHO type B2 thymomas and thymic carcinomas (p<0.001). HMGB1 nuclear staining was strongest in A and AB, and gradually less in B1 = B2>B3>thymic carcinoma (p<0.001). Conversely, HMGB1 cytoplasmic staining intensities were as follows: A and AB (none), B1 (strong), B2 (moderate), B3 and thymic carcinoma (weak); (p<0.001). Fetal thymic tissue showed a distinct expression of RAGE and HMGB1 in subcapsular cortical epithelial cells which was found in 50% of myasthenic patients. Furthermore RAGE and HMGB1 were expressed in thymocytes, macrophages, Hassall's corpuscles, thymic medulla, and germinal center cells in myasthenic patients. Immunohistochemistry results were complemented by systemic measurements (immunosorbent assay): serum levels of soluble RAGE were significantly reduced in patients with epithelial tumors (p = 0.008); and in invasive tumors (p = 0.008). Whereas RAGE was equally reduced in thymic hyperplasia and epithelial tumors (p = 0.003), HMGB1 was only elevated in malignancies (p = 0.036). Results were most pronounced in thymic carcinomas. Thus, RAGE and HMGB1 are involved in the (patho-)physiology of thymus, as evidenced by differentiated thymic and systemic expression patterns that may act as diagnostic or therapeutic targets in autoimmune disease and cancer.
Collapse
Affiliation(s)
- Bernhard Moser
- Department of Thoracic Surgery, Division of Surgery, Medical University Vienna, Vienna, Austria
- * E-mail:
| | - Stefan Janik
- Department of Thoracic Surgery, Division of Surgery, Medical University Vienna, Vienna, Austria
| | | | | | - Christine Bekos
- Department of Thoracic Surgery, Division of Surgery, Medical University Vienna, Vienna, Austria
- Christian Doppler Laboratory for the Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria
| | - Anke Scharrer
- Department of Pathology, Medical University Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University Vienna, Vienna, Austria
| | - Ferenc Rényi-Vámos
- Department of General and Thoracic Surgery, National Institute of Oncology, Budapest, Hungary
| | - Walter Klepetko
- Department of Thoracic Surgery, Division of Surgery, Medical University Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Division of Surgery, Medical University Vienna, Vienna, Austria
- Christian Doppler Laboratory for the Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria
| |
Collapse
|
50
|
Li RL, Zhang ZZ, Peng M, Wu Y, Zhang JJ, Wang CY, Wang YL. Postoperative impairment of cognitive function in old mice: a possible role for neuroinflammation mediated by HMGB1, S100B, and RAGE. J Surg Res 2013; 185:815-24. [DOI: 10.1016/j.jss.2013.06.043] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 06/05/2013] [Accepted: 06/20/2013] [Indexed: 01/01/2023]
|