1
|
Cai M, Yang EJ. Effect of Combined Electroacupuncture and Selegiline Treatment in Alzheimer's Disease: An Animal Model. Front Pharmacol 2020; 11:606480. [PMID: 33362561 PMCID: PMC7758426 DOI: 10.3389/fphar.2020.606480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/10/2020] [Indexed: 11/13/2022] Open
Abstract
The complexity of pathological mechanisms in Alzheimer's disease (AD) poses significant challenges to the development of corresponding drugs. Symptom-specific pharmacological interventions and alternative treatments provide promising treatment possibilities. Therefore, we considered a combination of selegiline (SEL) and electroacupuncture (EA). We used an animal model with AD to investigate the effect of a combination of these treatments on cognitive function. 5XFAD mice received a week of SEL treatment and 2 weeks of EA. Novel object recognition and Y-maze tests were subsequently performed to assess their cognitive functions. To determine the molecular action of the combination treatment, Western blots, Aβ1-42 enzyme-linked immunosorbent assays (ELISA), and micro-positron-emission tomography were also performed to assess pathological markers and processes. The results were assessed based on the difference between untreated transgenic, SEL-treated, and SEL- and EA-treated groups of mice. Mice in the combined treatment group demonstrated significantly better cognitive functions, and lesser neuroinflammation than the comparative groups. In addition, mice treated with a combination of SEL and EA did not demonstrate a direct modulation of insoluble Aβ but demonstrated greater glucose metabolism. Our findings demonstrated that SEL combined with EA treatment was associated with better cognitive functioning due to inhibition of neuroinflammation and increased glucose metabolism relative to the comparative groups in a mouse model with AD.
Collapse
Affiliation(s)
- Mudan Cai
- Department of Herbal Medicine Research, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Eun Jin Yang
- Department of Clinical Research, Korea Institute of Oriental Medicine, Daejeon, South Korea
| |
Collapse
|
2
|
Yang EJ, Mahmood U, Kim H, Choi M, Choi Y, Lee JP, Chang MJ, Kim HS. Alterations in protein phosphorylation in the amygdala of the 5XFamilial Alzheimer's disease animal model. J Pharmacol Sci 2017; 133:261-267. [PMID: 28408165 DOI: 10.1016/j.jphs.2017.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/13/2017] [Accepted: 03/21/2017] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease is the most common disease underlying dementia in humans. Two major neuropathological hallmarks of AD are neuritic plaques primarily composed of amyloid beta peptide and neurofibrillary tangles primarily composed of hyperphosphorylated tau. In addition to impaired memory function, AD patients often display neuropsychiatric symptoms and abnormal emotional states such as confusion, delusion, manic/depressive episodes and altered fear status. Brains from AD patients show atrophy of the amygdala which is involved in fear expression and emotional processing as well as hippocampal atrophy. However, which molecular changes are responsible for the altered emotional states observed in AD remains to be elucidated. Here, we observed that the fear response as assessed by evaluating fear memory via a cued fear conditioning test was impaired in 5XFamilial AD (5XFAD) mice, an animal model of AD. Compared to wild-type mice, 5XFAD mice showed changes in the phosphorylation of twelve proteins in the amygdala. Thus, our study provides twelve potential protein targets in the amygdala that may be responsible for the impairment in fear memory in AD.
Collapse
Affiliation(s)
- Eun-Jeong Yang
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Usman Mahmood
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Hyunju Kim
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Moonseok Choi
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Yunjung Choi
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Jean-Pyo Lee
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, 1430 Tulane Ave, SL99, New Orleans, LA 70112, USA.
| | - Moon-Jeong Chang
- Department of Foods and Nutrition, College of Natural Science, Kookmin University, Seoul, Republic of Korea.
| | - Hye-Sun Kim
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea; Seoul National University College of Medicine, Bundang Hospital, Bundang-Gu, Sungnam, Republic of Korea; Neuroscience Research Institute, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW We reviewed clinical trials on active and passive anti-β-amyloid (Aβ) immunotherapy for the treatment of Alzheimer's disease with a particular focus on monoclonal antibodies against Aβ. RECENT FINDINGS Studies on anti-Alzheimer's disease immunotherapy published in the period from January 2012 to October 2013 were reviewed. SUMMARY Both active and passive anti-Aβ immunotherapies were shown to clear brain Aβ deposits. However, an active anti-Aβ vaccine (AN1792) has been discontinued because it caused meningoencephalitis in 6% of Alzheimer's disease patients treated. Among passive immunotherapeutics, two Phase III clinical trials in mild-to-moderate Alzheimer's disease patients with bapineuzumab, a humanized monoclonal antibody directed at the N-terminal sequence of Aβ, were disappointing. Another antibody, solanezumab, directed at the mid-region of Aβ, failed in two Phase III clinical trials in mild-to-moderate Alzheimer's disease patients. A third Phase III study with solanezumab is ongoing in mildly affected Alzheimer's disease patients based on encouraging results in this subgroup of patients. Second-generation active Aβ vaccines (ACC-001, CAD106, and Affitope AD02) and new passive anti-Aβ immunotherapies (gantenerumab and crenezumab) are being tested in prodromal Alzheimer's disease patients, in presymptomatic individuals with Alzheimer's disease-related mutations, or in asymptomatic individuals at risk of developing Alzheimer's disease to definitely test the Aβ cascade hypothesis of Alzheimer's disease.
Collapse
|
4
|
Panza F, Solfrizzi V, Imbimbo BP, Tortelli R, Santamato A, Logroscino G. Amyloid-based immunotherapy for Alzheimer's disease in the time of prevention trials: the way forward. Expert Rev Clin Immunol 2014; 10:405-19. [DOI: 10.1586/1744666x.2014.883921] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
5
|
Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, Logroscino G, Santamato A, Greco A, Seripa D, Pilotto A. Immunotherapy for Alzheimer's disease: from anti-β-amyloid to tau-based immunization strategies. Immunotherapy 2012; 4:213-38. [PMID: 22339463 DOI: 10.2217/imt.11.170] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The exact mechanisms leading to Alzheimer's disease (AD) are largely unknown, limiting the identification of effective disease-modifying therapies. The two principal neuropathological hallmarks of AD are extracellular β-amyloid (Aβ), peptide deposition (senile plaques) and intracellular neurofibrillary tangles containing hyperphosphorylated tau protein. During the last decade, most of the efforts of the pharmaceutical industry were directed against the production and accumulation of Aβ. The most innovative of the pharmacological approaches was the stimulation of Aβ clearance from the brain of AD patients via the administration of Aβ antigens (active vaccination) or anti-Aβ antibodies (passive vaccination). Several active and passive anti-Aβ vaccines are under clinical investigation. Unfortunately, the first active vaccine (AN1792, consisting of preaggregate Aβ and an immune adjuvant, QS-21) was abandoned because it caused meningoencephalitis in approximately 6% of treated patients. Anti-Aβ monoclonal antibodies (bapineuzumab and solanezumab) are now being developed. The clinical results of the initial studies with bapineuzumab were equivocal in terms of cognitive benefit. The occurrence of vasogenic edema after bapineuzumab, and more rarely brain microhemorrhages (especially in Apo E ε4 carriers), has raised concerns on the safety of these antibodies directed against the N-terminus of the Aβ peptide. Solanezumab, a humanized anti-Aβ monoclonal antibody directed against the midregion of the Aβ peptide, was shown to neutralize soluble Aβ species. Phase II studies showed a good safety profile of solanezumab, while studies on cerebrospinal and plasma biomarkers documented good signals of pharmacodynamic activity. Although some studies suggested that active immunization may be effective against tau in animal models of AD, very few studies regarding passive immunization against tau protein are currently available. The results of the large, ongoing Phase III trials with bapineuzumab and solanezumab will tell us if monoclonal anti-Aβ antibodies may slow down the rate of deterioration of AD. Based on the new diagnostic criteria of AD and on recent major failures of anti-Aβ drugs in mild-to-moderate AD patients, one could argue that clinical trials on potential disease-modifying drugs, including immunological approaches, should be performed in the early stages of AD.
Collapse
Affiliation(s)
- Francesco Panza
- Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, Foggia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Armstrong RA. The pathogenesis of Alzheimer's disease: a reevaluation of the "amyloid cascade hypothesis". Int J Alzheimers Dis 2011; 2011:630865. [PMID: 21331369 PMCID: PMC3038555 DOI: 10.4061/2011/630865] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 01/04/2011] [Indexed: 12/27/2022] Open
Abstract
The most influential theory to explain the pathogenesis of Alzheimer's disease (AD) has been the "Amyloid Cascade Hypothesis" (ACH) first formulated in 1992. The ACH proposes that the deposition of β-amyloid (Aβ) is the initial pathological event in AD leading to the formation of senile plaques (SPs) and then to neurofibrillary tangles (NFTs) death of neurons, and ultimately dementia. This paper examines two questions regarding the ACH: (1) is there a relationship between the pathogenesis of SPs and NFTs, and (2) what is the relationship of these lesions to disease pathogenesis? These questions are examined in relation to studies of the morphology and molecular determinants of SPs and NFTs, the effects of gene mutation, degeneration induced by head injury, the effects of experimentally induced brain lesions, transgenic studies, and the degeneration of anatomical pathways. It was concluded that SPs and NFTs develop independently and may be the products rather than the causes of neurodegeneration in AD. A modification to the ACH is proposed which may better explain the pathogenesis of AD, especially of late-onset cases of the disease.
Collapse
|
7
|
Liang WS, Dunckley T, Beach TG, Grover A, Mastroeni D, Ramsey K, Caselli RJ, Kukull WA, McKeel D, Morris JC, Hulette CM, Schmechel D, Reiman EM, Rogers J, Stephan DA. Neuronal gene expression in non-demented individuals with intermediate Alzheimer's Disease neuropathology. Neurobiol Aging 2008; 31:549-66. [PMID: 18572275 DOI: 10.1016/j.neurobiolaging.2008.05.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Revised: 05/02/2008] [Accepted: 05/06/2008] [Indexed: 12/22/2022]
Abstract
While the clinical and neuropathological characterization of Alzheimer's Disease (AD) is well defined, our understanding of the progression of pathologic mechanisms in AD remains unclear. Post-mortem brains from individuals who did not fulfill clinical criteria for AD may still demonstrate measurable levels of AD pathologies to suggest that they may have presented with clinical symptoms had they lived longer or are able to stave off disease progression. Comparison between such individuals and those clinically diagnosed and pathologically confirmed to have AD will be key in delineating AD pathogenesis and neuroprotection. In this study, we expression profiled laser capture microdissected non-tangle bearing neurons in 6 post-mortem brain regions that are differentially affected in the AD brain from 10 non-demented individuals demonstrating intermediate AD neuropathologies (NDAD; Braak stage of II through IV and CERAD rating of moderate to frequent) and evaluated this data against that from individuals who have been diagnosed with late onset AD as well as healthy elderly controls. We identified common statistically significant expression changes in both NDAD and AD brains that may establish a degenerative link between the two cohorts, in addition to NDAD specific transcriptomic changes. These findings pinpoint novel targets for developing earlier diagnostics and preventative therapies for AD prior to diagnosis of probable AD. We also provide this high-quality, low post-mortem interval (PMI), cell-specific, and region-specific NDAD/AD reference data set to the community as a public resource.
Collapse
Affiliation(s)
- Winnie S Liang
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Li B, Chohan MO, Grundke-Iqbal I, Iqbal K. Disruption of microtubule network by Alzheimer abnormally hyperphosphorylated tau. Acta Neuropathol 2007; 113:501-11. [PMID: 17372746 PMCID: PMC3191942 DOI: 10.1007/s00401-007-0207-8] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2006] [Revised: 02/09/2007] [Accepted: 02/09/2007] [Indexed: 01/06/2023]
Abstract
Hyperphosphorylated tau has long been proposed as the key molecule disrupting normal neuronal microtubule dynamics and leading to neurofibrillary degeneration in Alzheimer disease. Here we provide a direct evidence of hyperphosphorylated tau-induced disruption of microtubule network. Using Nocodozole-treated and detergent-extracted cells, we created a neuronal environment in mouse embryonic fibroblasts, 3T3 cells, by replacing their cytoplasm with adult rat brain cytosol. By recreating neuronal microtubule network in these cells, we were able to follow the effects of hyperphosphorylated tau on microtubule dynamics in real time. Whereas recombinant human brain tau promoted assembly and bundling of microtubules, abnormally hyperphosphorylated tau isolated from Alzheimer disease brain cytosol (AD P-tau) inhibited the assembly and disrupted preformed microtubule network by sequestering normal brain tau and MAP2. This breakdown of the microtubule network was reversed by treatment of the extracted cells with protein phosphatase-2A. This study, for the first time, provides direct mechanistic insights into the molecular basis of both axonal and dendritic neurodegeneration seen in Alzheimer disease.
Collapse
|
9
|
Barrachina M, Castaño E, Ferrer I. TaqMan PCR assay in the control of RNA normalization in human post-mortem brain tissue. Neurochem Int 2006; 49:276-84. [PMID: 16522342 DOI: 10.1016/j.neuint.2006.01.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2005] [Revised: 01/20/2006] [Accepted: 01/23/2006] [Indexed: 10/24/2022]
Abstract
The brain tissue obtained after death is subjected to several circumstances that can affect RNA integrity. The present study has been directed to reveal possible pitfalls and to control RNA normalization in post-mortem samples in order to recognize the limitations and minimize errors when using TaqMan PCR technology. This has been carried out in samples of the frontal cortex in a series of control and diseased cases covering Parkinson's disease, dementia with Lewy bodies pure form and common form, and Alzheimer's disease. Special attention has been paid to the value of the agonal state, post-mortem delay and pH of the nervous tissue as approximate predictors of the quality of RNA, as well as to the use of the Bioanalyzer to confirm RNA preservation. In addition, since possible disease-modified mRNAs have to be normalized with ideal unaltered RNAs, TaqMan human endogenous control plates have been used to determine the endogenous control most appropriate for the study. beta-glucuronidase (GUS) and beta-actin were good endogenous controls because their expression levels showed a small variation across a representative number of control and pathological cases. RNA stability was also analysed in a paradigm mimicking cumulative delay in tissue processing. GUS mRNA levels were not modified although beta-actin mRNA levels showed degradation at 22 h. Finally, the control of RNA degradation for the normalization of genes of interest was also tested. mRNA expression levels for superoxide dismutase 1 (SOD1) and metalloproteinase domain 22 (ADAM22) were examined at several artificial post-mortem times, and their expression levels compared with those for putative controls beta-actin and GUS. In our paradigm, the expressions of SOD1 and ADAM22 were apparently not modified when normalized with beta-actin. Yet their expression levels were reduced with post-mortem delay when values were normalized with GUS. Taken together, these observations point to practical consequences in TaqMan PCR studies. Short post-mortem delays and acceptable pH of the brain are not sufficient to rule out RNA degradation. The selection of adequate endogenous controls is pivotal in the study. beta-actin and GUS are found to be good endogenous controls in these pathologies, although GUS but not beta-actin expression levels are preserved in samples with long post-mortem delay.
Collapse
Affiliation(s)
- Marta Barrachina
- Institut de Neuropatologia, Servei d'Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | | | | |
Collapse
|
10
|
Hu WP, Chang GL, Chen SJ, Kuo YM. Kinetic analysis of beta-amyloid peptide aggregation induced by metal ions based on surface plasmon resonance biosensing. J Neurosci Methods 2006; 154:190-7. [PMID: 16457893 DOI: 10.1016/j.jneumeth.2005.12.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2005] [Revised: 12/15/2005] [Accepted: 12/19/2005] [Indexed: 11/28/2022]
Abstract
Recent studies suggest that beta-amyloid (Abeta) aggregation and toxicity are facilitated by metal ions. This study aims to evaluate the kinetics of Abeta aggregation/dissociation in the presence of metal ions and to investigate the efficacy of a metal chelator to disrupt the metal ion-induced Abeta aggregates. Soluble Abeta(1-40) peptide was immobilized on a surface plasmon resonance biosensing surface and aggregation induced by contact with soluble Abeta with or without metal ions. Our study revealed that all the tested metal ions promoted Abeta aggregation but with different kinetics. Among them, Cu(II) ions had the highest association constant, and reached the maximum binding in 10 min. However, the Cu(II)-induced Abeta aggregates were unstable. Other ions attained the maximum Abeta binding at much longer times: 45 min for Ca(II), 60 min for Fe(II), Fe(III), and Zn(II) ions. The Abeta aggregates induced by Fe(III) ions had the greatest stability. The metal ion-induced Abeta(1-40) aggregates could be disrupted by the metal chelator, EDTA, suggesting a metal chelator may serve as a pharmacological agent to interfere with Abeta aggregation. Finally, this study demonstrates that the SPR biosensor can be an effective and efficient setup to investigate the mechanism of Abeta aggregation.
Collapse
Affiliation(s)
- Wen-Pin Hu
- Institute of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | | | | | | |
Collapse
|
11
|
Chung CW, Song YH, Kim IK, Yoon WJ, Ryu BR, Jo DG, Woo HN, Kwon YK, Kim HH, Gwag BJ, Mook-Jung IH, Jung YK. Proapoptotic effects of tau cleavage product generated by caspase-3. Neurobiol Dis 2001; 8:162-72. [PMID: 11162250 DOI: 10.1006/nbdi.2000.0335] [Citation(s) in RCA: 165] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Using an in vitro translation assay to screen a human brain cDNA library, we isolated the microtubule-associated protein Tau and determined it to be a caspase-3 substrate whose C-terminal cleavage occurred during neuronal apoptosis. DeltaTau, the 50-kDa cleavage product, was detected by Western blot in apoptotic cortical cells probed with anti-PHF-1 and anti-Tau-5 antibodies, but not anti-T-46 antibody which recognizes the C-terminus. Overexpression of DeltaTau in SK-N-BE2(C) cells significantly increased the incidence of cell death. Staurosporine-induced Tau cleavage was blocked by 20 microM z-Asp-Glu-Val-Asp-chloromethylketone, a caspase-3 inhibitor, and in vitro, Tau was selectively cleaved by caspase-3 or calpain, a calcium-activated protease, but not by caspases-1, -8, or -9. (D421E)-Tau, a mutant in which Asp421 was replaced with a Glu, was resistant to cleavage by caspase-3 and tended to suppress staurosporine-induced cell death more efficiently than did wild-type Tau in both transient and stable expression systems. Finally, the incidence of DeltaTau-induced cell death was augmented by expression of Abeta precursor protein (APP) or Swedish APP mutant. Taken together, these results suggest that the caspase-3 cleavage product of Tau may contribute to the progression of neuronal cell death in Alzheimer's disease.
Collapse
Affiliation(s)
- C W Chung
- Department of Life Science, Kwangju Institute of Science and Technology, and Biomedical Brain Research Center, National Institute of Health, Puk-Gu, Kwangju, 500-712, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Oyama R, Yamamoto H, Titani K. Glutamine synthetase, hemoglobin alpha-chain, and macrophage migration inhibitory factor binding to amyloid beta-protein: their identification in rat brain by a novel affinity chromatography and in Alzheimer's disease brain by immunoprecipitation. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1479:91-102. [PMID: 11004532 DOI: 10.1016/s0167-4838(00)00057-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Proteins binding to amyloid beta-protein (Abeta) may modulate the accumulation of Abeta in Alzheimer's disease (AD) brain. We developed a monomeric Abeta column for isolation of the proteins binding to Abeta from rat brain. By amino acid sequence analysis and immunoreactivity with specific antibodies, we identified three new Abeta-binding proteins, glutamine synthetase, hemoglobin alpha-chain, and macrophage migration inhibitory factor as well as serum albumin, beta-tubulin, and glyceraldehyde-3-phosphate dehydrogenase already identified as proteins bound to amyloid beta-protein precursor. In addition, the retained fraction contained both apolipoprotein E and alpha(1)-antichymotrypsin already known as Abeta binding proteins. Furthermore, we detected the complexes of these new binding proteins with Abeta in a soluble fraction of the cerebral cortex of AD brain by immunoprecipitation. Our results suggest that these binding proteins also associate with Abeta, leading to the clearance or the accumulation of Abeta and the neuronal cell damage in human brain.
Collapse
Affiliation(s)
- R Oyama
- Department of Neuropathology, Faculty of Medicine, University of Tokyo, Japan
| | | | | |
Collapse
|
13
|
Tohgi H, Utsugisawa K, Nagane Y, Yoshimura M, Ukitsu M, Genda Y. The methylation status of cytosines in a tau gene promoter region alters with age to downregulate transcriptional activity in human cerebral cortex. Neurosci Lett 1999; 275:89-92. [PMID: 10568506 DOI: 10.1016/s0304-3940(99)00731-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Changes with age in the methylation status of cytosines in a promoter region of the tau gene were investigated in autopsy human cerebral cortex, using the bisulfite method, polymerase chain reaction (PCR) and direct sequencing of PCR products. While the total number of methylcytosines decreased with age, the changes in methylation status differed among transcription factor binding sites. Cytosines in the AP2-binding sites were never methylated in any of the cases studied at any age. Methylcytosines in the binding sites for Sp1, a transcriptional activator, significantly increased with age, whereas those in the binding sites for GCF, a repressor of GC-rich promoters, significantly decreased with age. These findings suggest that the methylation status of cytosines in the promoter region of the tau gene alters with age to decrease its transcriptional activity in the human cerebral cortex.
Collapse
Affiliation(s)
- H Tohgi
- Department of Neurology, Iwate Medical University, Morioka, Japan
| | | | | | | | | | | |
Collapse
|
14
|
Morihara T, Kudo T, Ikura Y, Kashiwagi Y, Miyamae Y, Nakamura Y, Tanaka T, Shinozaki K, Nishikawa T, Takeda M. Increased tau protein level in postmortem cerebrospinal fluid. Psychiatry Clin Neurosci 1998; 52:107-10. [PMID: 9682942 DOI: 10.1111/j.1440-1819.1998.tb00981.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Microtubule-associated protein tau has been reported to be significantly increased in cerebrospinal fluid (CSF) of the patients with Alzheimer's disease (AD), which suggests that it is possibly a biological marker for the diagnosis of AD. The underlying mechanism of the increased tau level in CSF, however, is not known. In this study, the tau levels were compared between antemortem and postmortem CSF. The postmortem tau levels in CSF were significantly increased in all groups including AD, neurological control, and nondemented control. A striking elevation of CSF tau was observed during the postmortem change with the nondemented subjects. These findings may offer some insight into the understanding of the mechanism of the increased tau level in CSF with AD and other related disorders.
Collapse
Affiliation(s)
- T Morihara
- Department of Neuropsychiatry, Osaka University Medical School, Suita City, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Chauhan NB, Lee JM, Siegel GJ. Na,K-ATPase mRNA levels and plaque load in Alzheimer's disease. J Mol Neurosci 1997; 9:151-66. [PMID: 9481617 DOI: 10.1007/bf02800498] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
UNLABELLED The expression of Na,K-ATPase alpha 1- and alpha 3-mRNAs was analyzed by in situ hybridization in the superior frontal cortex and cerebellum of brains from five Alzheimer's disease (AD), five nondemented age-matched, and three young control subjects. Brains with well-preserved RNA, tested by Northern hybridization of immobilized RNA with [32P]-labeled human beta-actin riboprobe, were chosen for analysis. In situ hybridization was performed on formalin-fixed, 5 microns-thick Paraplast sections with [35S]-labeled riboprobes prepared by in vitro transcription of the respective linearized clones: a 537-bp EcoRI-PstI fragment of alpha 1-cDNA and a 342-bp PstI-EcoRI fragment of alpha 3-cDNA. In cortex, grains related to mRNA were measured by density per unit area in five cortical columns separated by 1.0-1.2 cm in each of two adjacent sections. Each cortical column of 180-micron width was divided into four depths orthogonal to the pial surface between the pia and the white matter. Amyloid plaques were counted in the same regions of adjacent sections. In addition, alpha 3-mRNA grain clusters over individual pyramidal neurons within depth 4 were analyzed. We found the following significant changes (p < 0.05): 1. Increases in total alpha 1-mRNA by 13-19% in AD compared to young and by 7-12% in AD compared to age-matched controls. 2. Decrease in total alpha 3-mRNA by 31-38% in AD compared to young and age-matched controls. 3. Decrease in alpha 3-mRNA content over individual pyramidal perikarya by 14% in normal aged brains without plaques compared to young controls, and by 44% in AD relative to young controls and by 35% compared to age-matched controls. No significant difference (p < 0.2) was found with respect to alpha 1- or alpha 3-mRNA in cerebellar cortex or individual Purkinje cells among any of the groups. In addition, there was a trend toward an inverse correlation between the levels of alpha 3-mRNA and of diffuse plaques, but not of neuritic plaques, in AD cases. IN CONCLUSION 1. The increases in alpha 1-mRNA in AD may be related to an increased reactive gliosis. 2. The declines in alpha 3-mRNA per individual neuron found in normal aging occur prior to the formation of diffuse plaques and are greatly accelerated in AD. 3. The declines in alpha 3-mRNA per neuron found in normal aging may predispose to or potentiate AD pathogenesis.
Collapse
Affiliation(s)
- N B Chauhan
- Molecular and Cellular Neuroscience Laboratory, Edward Hines Jr. Veterans Affairs Hospital, IL 60141, USA
| | | | | |
Collapse
|
17
|
Oyama F, Shimada H, Oyama R, Ihara Y. Apolipoprotein E genotype, Alzheimer's pathologies and related gene expression in the aged population. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1995; 29:92-8. [PMID: 7770005 DOI: 10.1016/0169-328x(94)00233-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We have investigated the effect of genotypes of apolipoprotein E (ApoE) on the pathologies found in Alzheimer's disease (AD) and its related gene expression in 38 aged human brains obtained from consecutive autopsied cases. ApoE2/3, -3/3, -3/4, and -4/4 were typed in those aged brains, with ApoE3/3 being most prevalent. The AD pathologies were undetectable in ApoE2/3 brains, but were frequently observed in the other ApoE groups. In ApoE3/3 brains, 55%, 34%, and 24% of the cortical sections examined showed senile plaques (SPs), neurofibrillary tangles (NFTs), and cerebral amyloid angiopathy (CAA), respectively. In ApoE4/4 brains, the SP formation was significantly higher. The ApoE genotype neither affected ApoE, APP, or tau mRNA level, nor the differential expression of the latter two. These results suggest that ApoE4/4 accelerates and ApoE2/3 decelerates the development of the AD pathologies in the aged brain, but this is not through alterations of the APP and tau gene expression.
Collapse
Affiliation(s)
- F Oyama
- Department of Neuropathology, Faculty of Medicine, University of Tokyo, Japan
| | | | | | | |
Collapse
|
18
|
Choo LP, Mansfield JR, Pizzi N, Somorjai RL, Jackson M, Halliday WC, Mantsch HH. Infrared spectra of human central nervous system tissue: Diagnosis of alzheimer's disease by multivariate analyses. ACTA ACUST UNITED AC 1995. [DOI: 10.1002/bspy.350010208] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Dudek SM, Johnson GV. Transglutaminase facilitates the formation of polymers of the beta-amyloid peptide. Brain Res 1994; 651:129-33. [PMID: 7922559 DOI: 10.1016/0006-8993(94)90688-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
One of the major pathological characteristics of Alzheimer's disease is the increased number of amyloid-containing senile plaques within the brain. The dense cores of these plaques are composed primarily of highly insoluble aggregates of a 39-43-residue peptide referred to as the beta-amyloid peptide (beta A). The mechanisms by which these insoluble extracellular deposits of beta A are formed remain unknown. In this study, the cross-linking of beta A by the calcium-dependent enzyme, transglutaminase was examined. Transglutaminases are a family of enzymes which are found in brain, and catalyse the cross-linking of specific proteins into insoluble polymers. Synthetic beta A (1-40) was readily cross-linked by transglutaminase, forming multimers in a time-dependent fashion. Furthermore, a second peptide with a substitution similar to that in the Dutch-type hereditary amyloidosis mutation (Glu22 to Gln) was also found to be a substrate for transglutaminase. Since transglutaminase covalently cross-links proteins through glutamine residues, it is suggested that transglutaminase contributes to amyloid deposition in Dutch-type hereditary amyloidosis, and possibly Alzheimer's disease.
Collapse
Affiliation(s)
- S M Dudek
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham 35294-0017
| | | |
Collapse
|
20
|
Oyama F, Cairns NJ, Shimada H, Oyama R, Titani K, Ihara Y. Down's syndrome: up-regulation of beta-amyloid protein precursor and tau mRNAs and their defective coordination. J Neurochem 1994; 62:1062-6. [PMID: 8113792 DOI: 10.1046/j.1471-4159.1994.62031062.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Almost all patients > 40 years of age with Down's syndrome (DS) develop the pathology characteristic of Alzheimer's disease: abundant beta-amyloid plaques and neurofibrillary tangles. We have investigated the gene expression of beta-amyloid protein precursor (APP) and tau in DS and age-matched control brains and found that levels of both mRNAs were significantly elevated in DS. Such up-regulation was not observed in two other neuronal proteins. A correlation between total APP and tau mRNA levels was also found in DS brain but distinct from the pattern observed in normal brain. Although a proportionality existed between APP-695 mRNA and three-repeat tau mRNA in DS, the proportionality between APP-751 mRNA and four-repeat tau mRNA, which is normally present, was not observed. Thus, DS brains are primarily characterized by the up-regulation of tau mRNA as well as APP mRNA and disruption of the coordinate expression between APP-751 and four-repeat tau.
Collapse
Affiliation(s)
- F Oyama
- Division of Biomedical Polymer Science, School of Medicine, Fujita Health University, Aichi, Japan
| | | | | | | | | | | |
Collapse
|