1
|
Valverde-Salazar V, Ruiz-Gabarre D, García-Escudero V. Alzheimer's Disease and Green Tea: Epigallocatechin-3-Gallate as a Modulator of Inflammation and Oxidative Stress. Antioxidants (Basel) 2023; 12:1460. [PMID: 37507998 PMCID: PMC10376369 DOI: 10.3390/antiox12071460] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, characterised by a marked decline of both memory and cognition, along with pathophysiological hallmarks including amyloid beta peptide (Aβ) accumulation, tau protein hyperphosphorylation, neuronal loss and inflammation in the brain. Additionally, oxidative stress caused by an imbalance between free radicals and antioxidants is considered one of the main risk factors for AD, since it can result in protein, lipid and nucleic acid damage and exacerbate Aβ and tau pathology. To date, there is a lack of successful pharmacological approaches to cure or even ameliorate the terrible impact of this disease. Due to this, dietary compounds with antioxidative and anti-inflammatory properties acquire special relevance as potential therapeutic agents. In this context, green tea, and its main bioactive compound, epigallocatechin-3-gallate (EGCG), have been targeted as a plausible option for the modulation of AD. Specifically, EGCG acts as an antioxidant by regulating inflammatory processes involved in neurodegeneration such as ferroptosis and microglia-induced cytotoxicity and by inducing signalling pathways related to neuronal survival. Furthermore, it reduces tau hyperphosphorylation and aggregation and promotes the non-amyloidogenic route of APP processing, thus preventing the formation of Aβ and its subsequent accumulation. Taken together, these results suggest that EGCG may be a suitable candidate in the search for potential therapeutic compounds for neurodegenerative disorders involving inflammation and oxidative stress, including Alzheimer's disease.
Collapse
Affiliation(s)
- Víctor Valverde-Salazar
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Daniel Ruiz-Gabarre
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Vega García-Escudero
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, 28031 Madrid, Spain
- Institute for Molecular Biology-IUBM, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
2
|
Wenzel TJ, Nyarko JNK, Heistad RM, Pennington PR, Phenix CP, Mousseau DD. An (Immuno) Fluorescence Protocol for Monitoring Monoamine Oxidase A/B Protein Distribution Within the Cell. Methods Mol Biol 2023; 2558:143-161. [PMID: 36169861 DOI: 10.1007/978-1-0716-2643-6_11] [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: 06/16/2023]
Abstract
The influence of a protein is not determined exclusively by its level of expression, but also by its localization within the cell. The literature often refers to the enzyme monoamine oxidase (MAO) as a mitochondrial enzyme, yet there is evidence that mitochondria-independent pools of MAO exist. These pools of MAO could exert distinct influences across physiological as well as pathological phenotypes. Fluorescence microscopy is a powerful tool for spatially resolving target proteins in cell and tissue preparations. This can rely on an antibody-based probe that targets the endogenous protein, e.g., immunofluorescence. In the event that antibodies might not be readily available or if one is interested in characterizing a variant of the wild-type protein, then a recombinant protein with a fluorescent fusion "tag" is preferred. We now describe a protocol for the detection of endogenous MAO using indirect immunofluorescence and a version of the protocol with minor modification for detecting (green) fluorescent protein-tagged MAOs. One observation we can highlight using these easily adaptable approaches is that MAO A and MAO B do not follow similar patterns of distribution throughout the cell, suggesting potential expression of MAO A and MAO B on distinct pools of mitochondria. Furthermore, distinct subcellular compartmentalization is suggested by the fact that a pool of MAO A, but not MAO B, is associated with certain lysosomal compartments. However, directed and quantitative studies will be required before any definitive statement can be made on these intriguing possibilities.
Collapse
Affiliation(s)
- Tyler J Wenzel
- Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Ryan M Heistad
- Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Paul R Pennington
- Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Chris P Phenix
- Department of Chemistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Darrell D Mousseau
- Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
3
|
Smith AJ, Duan T, Verkman AS. Aquaporin-4 reduces neuropathology in a mouse model of Alzheimer's disease by remodeling peri-plaque astrocyte structure. Acta Neuropathol Commun 2019; 7:74. [PMID: 31068220 PMCID: PMC6506955 DOI: 10.1186/s40478-019-0728-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 04/23/2019] [Indexed: 01/01/2023] Open
Abstract
Redistribution of the water channel aquaporin-4 (AQP4) away from astrocyte endfeet and into parenchymal processes is a striking histological feature in mouse models of Alzheimer’s disease (AD) and other neurological conditions with prominent astrogliosis. AQP4 redistribution has been proposed to impair bulk Aβ clearance in AD, resulting in increased amyloid deposition in the brain; however, this finding is controversial. Here, we provide evidence in support of a different and novel role of AQP4 in AD. We found that Aqp4 deletion significantly increased amyloid deposition in cerebral cortex of 5xFAD mice, with an increase in the relative number of fibrillar vs. dense core plaques. AQP4 deficient 5xFAD mice also showed a significant reduction in the density of GFAP labeled peri-plaque astrocyte processes. Microglial plaque coverage was also significantly reduced, suggesting astrocyte involvement in organizing the peri-plaque glial response. The alterations in peri-plaque glial structure were accompanied by increased neuronal uptake of Aβ and an increase in the number of dystrophic neurites surrounding plaques. On the basis of these findings, we propose that redistribution of AQP4 into the parenchymal processes facilitates astrocyte structural plasticity and the formation of a reactive glial net around plaques that protects neurons from the deleterious effects of Aβ aggregates. AQP4 redistribution may thus facilitate plaque containment and reduce neuropathology in AD.
Collapse
|
4
|
Yuan Q, Zhang H, Zhang X, Huang P, Liu R, Song YQ, Wu W, Lin ZX. Association Between Axonopathy and Amyloid Plaques in the Spinal Cord of the Transgenic Mice of Alzheimer's Disease. Neuroscience 2019; 409:152-161. [PMID: 31034974 DOI: 10.1016/j.neuroscience.2019.04.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/12/2019] [Accepted: 04/16/2019] [Indexed: 11/29/2022]
Abstract
Axonopathy manifested by axon swellings might constitute one of the earliest pathological features of Alzheimer's disease. It has been proposed that axonopathy might be associated with the origin of Aβ plaques. However, how axonopathy leads to Aβ plaque pathogenesis remains elusive. Our previous studies have shown that Aβ neuropathology (mainly diffuse plaques) selectively occurred in the regions of corticospinal tract (CST) pathway and its innervated region in the spinal cord of TgCRND8 mice. In this study, we investigated the occurrence and progression of axonopathy and the possible implication in Aβ plaque pathogenesis in the spinal cord of TgCRND8 mice. By anterograde labeling of CST system with a neuroanatomical tracer, we found that dilated corticospinal axons started to appear at 7 months, then exhibited an age-dependent increase. These abnormal structures appear before any plaque deposits are visible in the spinal cord of the mice. Importantly, they colocalized with Aβ plaques in either the white matter or gray matter of the spinal cord at later stages, suggesting that these axonal swellings might represent the initial stages of Aβ plaque formation, and could play a role in Aβ plaque pathogenesis. Furthermore, using ultrastructural analysis we demonstrated that intracellular contents in the axonal dystrophies such as various dense vesicles leaked out into the extracellular matrix under a condition of axon swelling rupture in CST pathways of spinal cord. This provided precise structural evidence that how the Aβ leaks out from the axonal dystrophies into extracellular matrix and how an axonal swelling might serve as a nidus of amyloid plaque formation.
Collapse
Affiliation(s)
- Qiuju Yuan
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong; Brain Research Centre, Faculty of Medicine, The Chinese University of Hong Kong.
| | - Hongwei Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong
| | - Xie Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong
| | - Pengyun Huang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong
| | - Rong Liu
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - You-Qiang Song
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Wutian Wu
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong; GHM Institute of CNS regeneration, Jinan University, Guangzhou, China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong; Brain Research Centre, Faculty of Medicine, The Chinese University of Hong Kong.
| |
Collapse
|
5
|
Nyarko JNK, Quartey MO, Heistad RM, Pennington PR, Poon LJ, Knudsen KJ, Allonby O, El Zawily AM, Freywald A, Rauw G, Baker GB, Mousseau DD. Glycosylation States of Pre- and Post-synaptic Markers of 5-HT Neurons Differ With Sex and 5-HTTLPR Genotype in Cortical Autopsy Samples. Front Neurosci 2018; 12:545. [PMID: 30147642 PMCID: PMC6096231 DOI: 10.3389/fnins.2018.00545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/19/2018] [Indexed: 11/13/2022] Open
Abstract
The serotonin (5-hydroxytryptamine, 5-HT) transporter (5-HTT) gene-linked polymorphic region (5-HTTLPR) is thought to alter 5-HT signaling and contribute to behavioral and cognitive phenotypes in depression as well as Alzheimer disease (AD). We explored how well the short (S) and long (L) alleles of the 5-HTTLPR align with serotoninergic indices in 60 autopsied cortical samples from early-onset AD/EOAD and late-onset AD/LOAD donors, and age- and sex-matched controls. Stratifying data by either diagnosis-by-genotype or by sex-by-genotype revealed that the donor's 5-HTTLPR genotype, i.e., L/L, S/L, or S/S, did not affect 5-HTT mRNA or protein expression. However, the glycosylation of 5-HTT was significantly higher in control female (vs. male) samples and tended to decrease in female EOAD/LOAD samples, but remained unaltered in male LOAD samples. Glycosylated forms of the vesicular monoamine transporter (VMAT2) were lower in both male and female AD samples, while a sex-by-genotype stratification revealed a loss of VMAT2 glycosylation specifically in females with an L/L genotype. VMAT2 and 5-HTT glycosylation were correlated in male samples and inversely correlated in female samples in both stratification models. The S/S genotype aligned with lower levels of 5-HT turnover in females (but not males) and with an increased glycosylation of the post-synaptic 5-HT2C receptor. Interestingly, the changes in presynaptic glycosylation were evident primarily in female carriers of the APOE ε4 risk factor for AD. Our data do not support an association between 5-HTTLPR genotype and 5-HTT expression, but they do reveal a non-canonical association of 5-HTTLPR genotype with sex-dependent glycosylation changes in pre- and post-synaptic markers of serotoninergic neurons. These patterns of change suggest adaptive responses in 5-HT signaling and could certainly be contributing to the female prevalence in risk for either depression or AD.
Collapse
Affiliation(s)
- Jennifer N K Nyarko
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Maa O Quartey
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ryan M Heistad
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Paul R Pennington
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Lisa J Poon
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kaeli J Knudsen
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Odette Allonby
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Amr M El Zawily
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Andrew Freywald
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Gail Rauw
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Glen B Baker
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Darrell D Mousseau
- Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
6
|
Solleiro-Villavicencio H, Rivas-Arancibia S. Effect of Chronic Oxidative Stress on Neuroinflammatory Response Mediated by CD4 +T Cells in Neurodegenerative Diseases. Front Cell Neurosci 2018; 12:114. [PMID: 29755324 PMCID: PMC5934485 DOI: 10.3389/fncel.2018.00114] [Citation(s) in RCA: 290] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/11/2018] [Indexed: 12/16/2022] Open
Abstract
In a state of oxidative stress, there is an increase of reactive species, which induce an altered intracellular signaling, leading to dysregulation of the inflammatory response. The inability of the antioxidant defense systems to modulate the proinflammatory response is key to the onset and progression of neurodegenerative diseases. The aim of this work is to review the effect of the state of oxidative stress on the loss of regulation of the inflammatory response on the microglia and astrocytes, the induction of different CD4+T cell populations in neuroinflammation, as well as its role in some neurodegenerative diseases. For this purpose, an intentional search of original articles, short communications, and reviews, was carried out in the following databases: PubMed, Scopus, and Google Scholar. The articles reviewed included the period from 1997 to 2017. With the evidence obtained, we conclude that the loss of redox balance induces alterations in the differentiation and number of CD4+T cell subpopulations, leading to an increase in Th1 and Th17 response. This contributes to the development of neuroinflammation as well as loss of the regulation of the inflammatory response in neurodegenerative diseases such as Alzheimer's (AD), Parkinson's (PD), and Multiple Sclerosis (MS). In contrast, regulatory T cells (Tregs) and Th2 modulate the inflammatory response of effect of T cells, microglia, and astrocytes. In this respect, it has been found that the mobilization of T cells with anti-inflammatory characteristics toward damaged regions of the CNS can provide neuroprotection and become a therapeutic strategy to control inflammatory processes in neurodegeneration.
Collapse
Affiliation(s)
- Helena Solleiro-Villavicencio
- Laboratorio de Estrés Oxidativo y Plasticidad Cerebral, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| | - Selva Rivas-Arancibia
- Laboratorio de Estrés Oxidativo y Plasticidad Cerebral, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Ciudad de Mexico, Mexico
| |
Collapse
|
7
|
Cong Q, Jia H, Li P, Qiu S, Yeh J, Wang Y, Zhang ZL, Ao J, Li B, Liu H. p38α MAPK regulates proliferation and differentiation of osteoclast progenitors and bone remodeling in an aging-dependent manner. Sci Rep 2017; 7:45964. [PMID: 28382965 PMCID: PMC5382695 DOI: 10.1038/srep45964] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/07/2017] [Indexed: 02/07/2023] Open
Abstract
Bone mass is determined by the balance between bone formation, carried out by mesenchymal stem cell-derived osteoblasts, and bone resorption, carried out by monocyte-derived osteoclasts. Here we investigated the potential roles of p38 MAPKs, which are activated by growth factors and cytokines including RANKL and BMPs, in osteoclastogenesis and bone resorption by ablating p38α MAPK in LysM+monocytes. p38α deficiency promoted monocyte proliferation but regulated monocyte osteoclastic differentiation in a cell-density dependent manner, with proliferating p38α−/− cultures showing increased differentiation. While young mutant mice showed minor increase in bone mass, 6-month-old mutant mice developed osteoporosis, associated with an increase in osteoclastogenesis and bone resorption and an increase in the pool of monocytes. Moreover, monocyte-specific p38α ablation resulted in a decrease in bone formation and the number of bone marrow mesenchymal stem/stromal cells, likely due to decreased expression of PDGF-AA and BMP2. The expression of PDGF-AA and BMP2 was positively regulated by the p38 MAPK-Creb axis in osteoclasts, with the promoters of PDGF-AA and BMP2 having Creb binding sites. These findings uncovered the molecular mechanisms by which p38α MAPK regulates osteoclastogenesis and coordinates osteoclastogenesis and osteoblastogenesis.
Collapse
Affiliation(s)
- Qian Cong
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Jia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China.,Department of Biochemistry and Molecular cellular Biology Shanghai Jiaotong University, School of Medicine, 280 Chongqing Rd, Shanghai, 200025, China
| | - Ping Li
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shoutao Qiu
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - James Yeh
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yibin Wang
- Division of Molecular Medicine, Departments of Anesthesiology, Medicine and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine, Los Angeles, CA90095, USA
| | - Zhen-Lin Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Junping Ao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Baojie Li
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huijuan Liu
- Dept. of Osteoporosis and Bone Diseases, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
8
|
Compromised MAPK signaling in human diseases: an update. Arch Toxicol 2015; 89:867-82. [PMID: 25690731 DOI: 10.1007/s00204-015-1472-2] [Citation(s) in RCA: 754] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 02/09/2015] [Indexed: 02/08/2023]
Abstract
The mitogen-activated protein kinases (MAPKs) in mammals include c-Jun NH2-terminal kinase (JNK), p38 MAPK, and extracellular signal-regulated kinase (ERK). These enzymes are serine-threonine protein kinases that regulate various cellular activities including proliferation, differentiation, apoptosis or survival, inflammation, and innate immunity. The compromised MAPK signaling pathways contribute to the pathology of diverse human diseases including cancer and neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. The JNK and p38 MAPK signaling pathways are activated by various types of cellular stress such as oxidative, genotoxic, and osmotic stress as well as by proinflammatory cytokines such as tumor necrosis factor-α and interleukin 1β. The Ras-Raf-MEK-ERK signaling pathway plays a key role in cancer development through the stimulation of cell proliferation and metastasis. The p38 MAPK pathway contributes to neuroinflammation mediated by glial cells including microglia and astrocytes, and it has also been associated with anticancer drug resistance in colon and liver cancer. We here summarize recent research on the roles of MAPK signaling pathways in human diseases, with a focus on cancer and neurodegenerative conditions.
Collapse
|
9
|
Yuan Q, Su H, Zhang Y, Chau WH, Ng CT, Wu W, Lin ZX. Existence of different types of senile plaques between brain and spinal cord of TgCRND8 mice. Neurochem Int 2013; 62:211-20. [PMID: 23333593 DOI: 10.1016/j.neuint.2013.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 01/02/2013] [Accepted: 01/06/2013] [Indexed: 01/11/2023]
Abstract
Conflicting findings exist regarding the formation of diffuse and dense-core β-amyloid (Aβ) plaques in Alzheimer's disease (AD). In the present study, we characterized Aβ plaque types in the brain and spinal cord of TgCRND8 mice, which express a transgene incorporating both the Indiana mutation (V717F) and the Swedish mutations (K670N/M671L) in the human amyloid-β protein precursor (APP) gene. By combining immunohistochemistry and thioflavin S staining, we were able to define dense-core and diffuse plaques in neocortex of the brain and spinal cord of 9 week-, 5 month-, 10 month- and 20-month-old TgCRND8 mice. The senile plaques in the neocortex were predominantly dense-core plaques, even in the youngest mice. However, diffuse plaques were instead detected in spinal cord of the mice, regardless of age. Our results that relative predominance of dense-core plaques in the neocortex and diffuse plaques in the spinal cord of TgCNRD8 mice of all disease durations argue against the notion that diffuse plaques may represent an early stage in the evolution of dense-core plaques. Furthermore, we also found that the ratio of Aβ42/Aβ40 of the brain was much higher than that of the spinal cord by Aβ ELISA assay. Our findings strongly indicate that diffuse and dense-core plaques may form via independent processes in AD and Aβ42 is more prone to form dense-core plaques than is Aβ40.
Collapse
Affiliation(s)
- Qiuju Yuan
- School of Chinese Medicine, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | | | | | | | | | | | | |
Collapse
|
10
|
Jurk D, Wang C, Miwa S, Maddick M, Korolchuk V, Tsolou A, Gonos ES, Thrasivoulou C, Jill Saffrey M, Cameron K, von Zglinicki T. Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell 2012; 11:996-1004. [PMID: 22882466 PMCID: PMC3533793 DOI: 10.1111/j.1474-9726.2012.00870.x] [Citation(s) in RCA: 401] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In senescent cells, a DNA damage response drives not only irreversible loss of replicative capacity but also production and secretion of reactive oxygen species (ROS) and bioactive peptides including pro-inflammatory cytokines. This makes senescent cells a potential cause of tissue functional decline in aging. To our knowledge, we show here for the first time evidence suggesting that DNA damage induces a senescence-like state in mature postmitotic neurons in vivo. About 40–80% of Purkinje neurons and 20–40% of cortical, hippocampal and peripheral neurons in the myenteric plexus from old C57Bl/6 mice showed severe DNA damage, activated p38MAPkinase, high ROS production and oxidative damage, interleukin IL-6 production, heterochromatinization and senescence-associated β-galactosidase activity. Frequencies of these senescence-like neurons increased with age. Short-term caloric restriction tended to decrease frequencies of positive cells. The phenotype was aggravated in brains of late-generation TERC−/− mice with dysfunctional telomeres. It was fully rescued by loss of p21(CDKN1A) function in late-generation TERC−/−CDKN1A−/− mice, indicating p21 as the necessary signal transducer between DNA damage response and senescence-like phenotype in neurons, as in senescing fibroblasts and other proliferation-competent cells. We conclude that a senescence-like phenotype is possibly not restricted to proliferation-competent cells. Rather, dysfunctional telomeres and/or accumulated DNA damage can induce a DNA damage response leading to a phenotype in postmitotic neurons that resembles cell senescence in multiple features. Senescence-like neurons might be a source of oxidative and inflammatory stress and a contributor to brain aging.
Collapse
Affiliation(s)
- Diana Jurk
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Chunfang Wang
- Department of Life Sciences, The Open University, Milton Keynes MK7 6AA, UK
| | - Satomi Miwa
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Mandy Maddick
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Viktor Korolchuk
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Avgi Tsolou
- National Hellenic Research Foundation, Institute of Biological Research and Biotechnology, Athens, Greece
| | - Efstathios S. Gonos
- National Hellenic Research Foundation, Institute of Biological Research and Biotechnology, Athens, Greece
| | | | - M. Jill Saffrey
- Department of Life Sciences, The Open University, Milton Keynes MK7 6AA, UK
| | - Kerry Cameron
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Thomas von Zglinicki
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|