1
|
Wang YC, Huang APH, Yuan SP, Huang CY, Wu CC, Poly TN, Atique S, Kung WM. Association between Anemia and Risk of Parkinson Disease. Behav Neurol 2021; 2021:8360627. [PMID: 34306250 PMCID: PMC8279865 DOI: 10.1155/2021/8360627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/19/2021] [Indexed: 02/08/2023] Open
Abstract
METHODS We systematically searched articles on electronic databases such as PubMed, Embase, Scopus, and Google Scholar between January 1, 2000 and July 30, 2020. Articles were independently evaluated by two authors. We included observational studies (case-control and cohort) and calculated the risk ratios (RRs) for associated with anemia and PD. Heterogeneity among the studies was assessed using the Q and I 2 statistic. We utilized the random-effect model to calculate the overall RR with 95% CI. RESULTS A total of 342 articles were identified in the initial searches, and 7 full-text articles were evaluated for eligibility. Three articles were further excluded for prespecified reasons including insufficient data and duplications, and 4 articles were included in our systematic review and meta-analysis. A random effect model meta-analysis of all 4 studies showed no increased risk of PD in patients with anemia (N = 4, RRadjusted = 1.17 (95% CI: 0.94-1.45, p = 0.15). However, heterogeneity among the studies was significant (I 2 = 92.60, p = <0.0001). The pooled relative risk of PD in female patients with anemia was higher (N = 3, RRadjusted = 1.14 (95% CI: 0.83-1.57, p = 0.40) as compared to male patients with anemia (N = 3, RRadjusted = 1.09 (95% CI: 0.83-1.42, p = 0.51). CONCLUSION This is the first meta-analysis that shows that anemia is associated with higher risk of PD when compared with patients without anemia. However, more studies are warranted to evaluate the risk of PD among patients with anemia.
Collapse
Affiliation(s)
- Yao-Chin Wang
- Department of Emergency, Min-Sheng General Hospital, Taoyuan, Taiwan
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Abel Po-Hao Huang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Sheng-Po Yuan
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Department of Otorhinolaryngology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Otorhinolaryngology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chu-Ya Huang
- Taiwan College of Healthcare Executives, Taipei, Taiwan
| | - Chieh-Chen Wu
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei, Taiwan
| | - Tahmina Nasrin Poly
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Suleman Atique
- Department of Health Informatics, College of Public Health and Health Informatics, University of Ha'il, Ha'il, Saudi Arabia
| | - Woon-Man Kung
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei, Taiwan
| |
Collapse
|
2
|
Wang T, Zhang J, Xu Y. Epigenetic Basis of Lead-Induced Neurological Disorders. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17134878. [PMID: 32645824 PMCID: PMC7370007 DOI: 10.3390/ijerph17134878] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Environmental lead (Pb) exposure is closely associated with pathogenesis of a range of neurological disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), attention deficit/hyperactivity disorder (ADHD), etc. Epigenetic machinery modulates neural development and activities, while faulty epigenetic regulation contributes to the diverse forms of CNS (central nervous system) abnormalities and diseases. As a potent epigenetic modifier, lead is thought to cause neurological disorders through modulating epigenetic mechanisms. Specifically, increasing evidence linked aberrant DNA methylations, histone modifications as well as ncRNAs (non-coding RNAs) with AD cases, among which circRNA (circular RNA) stands out as a new and promising field for association studies. In 23-year-old primates with developmental lead treatment, Zawia group discovered a variety of epigenetic changes relating to AD pathogenesis. This is a direct evidence implicating epigenetic basis in lead-induced AD animals with an entire lifespan. Additionally, some epigenetic molecules associated with AD etiology were also known to respond to chronic lead exposure in comparable disease models, indicating potentially interlaced mechanisms with respect to the studied neurotoxic and pathological events. Of note, epigenetic molecules acted via globally or selectively influencing the expression of disease-related genes. Compared to AD, the association of lead exposure with other neurological disorders were primarily supported by epidemiological survey, with fewer reports connecting epigenetic regulators with lead-induced pathogenesis. Some pharmaceuticals, such as HDAC (histone deacetylase) inhibitors and DNA methylation inhibitors, were developed to deal with CNS disease by targeting epigenetic components. Still, understandings are insufficient regarding the cause–consequence relations of epigenetic factors and neurological illness. Therefore, clear evidence should be provided in future investigations to address detailed roles of novel epigenetic factors in lead-induced neurological disorders, and efforts of developing specific epigenetic therapeutics should be appraised.
Collapse
Affiliation(s)
| | | | - Yi Xu
- Correspondence: ; Tel.: +86-183-2613-5046
| |
Collapse
|
3
|
Shen X, Xia L, Liu L, Jiang H, Shannahan J, Du Y, Zheng W. Altered clearance of beta-amyloid from the cerebrospinal fluid following subchronic lead exposure in rats: Roles of RAGE and LRP1 in the choroid plexus. J Trace Elem Med Biol 2020; 61:126520. [PMID: 32325398 PMCID: PMC7541561 DOI: 10.1016/j.jtemb.2020.126520] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/26/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023]
Abstract
Formation of amyloid plaques is the hallmark of Alzheimer's disease. Our early studies show that lead (Pb) exposure in PDAPP transgenic mice increases β-amyloid (Aβ) levels in the cerebrospinal fluid (CSF) and hippocampus, leading to the formation of amyloid plaques in mouse brain. Aβ in the CSF is regulated by the blood-CSF barrier (BCB) in the choroid plexus. However, the questions as to whether and how Pb exposure affected the influx and efflux of Aβ in BCB remained unknown. This study was conducted to investigate whether Pb exposure altered the Aβ efflux in the choroid plexus from the CSF to blood, and how Pb may affect the expression and subcellular translocation of two major Aβ transporters, i.e., the receptor for advanced glycation end-products (RAGE) and the low density lipoprotein receptor protein-1 (LRP1) in the choroid plexus. Sprague-Dawley rats received daily oral gavage at doses of 0, 14 (low-dose), and 27 (high-dose) mg Pb/kg as Pb acetate, 5 d/wk, for 4 or 8 wks. At the end of Pb exposure, a solution containing Aβ40 (2.5 μg/mL) was infused to rat brain via a cannulated internal carotid artery. Subchronic Pb exposure at both dose levels significantly increased Aβ levels in the CSF and choroid plexus (p < 0.05) by ELISA. Confocal data showed that 4-wk Pb exposures prompted subcellular translocation of RAGE from the choroidal cytoplasm toward apical microvilli. Furthermore, it increased the RAGE expression in the choroid plexus by 34.1 % and 25.1 % over the controls (p < 0.05) in the low- and high- dose groups, respectfully. Subchronic Pb exposure did not significantly affect the expression of LRP1; yet the high-dose group showed LRP1 concentrated along the basal lamina. The data from the ventriculo-cisternal perfusion revealed a significantly decreased efflux of Aβ40 from the CSF to blood via the blood-CSF barrier. Incubation of freshly dissected plexus tissues with Pb in artificial CSF supported a Pb effect on increased RAGE expression. Taken together, these data suggest that Pb accumulation in the choroid plexus after subchronic exposure reduces the clearance of Aβ from the CSF to blood by the choroid plexus, which, in turn, leads to an increase of Aβ in the CSF. Interaction of Pb with RAGE and LRP1 in choroidal epithelial cells may contribute to the altered Aβ transport by the blood-CSF barrier in brain ventricles.
Collapse
Affiliation(s)
- Xiaoli Shen
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; School of Public Health, Qingdao University, Qingdao, China.
| | - Li Xia
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Luqing Liu
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Hong Jiang
- Departments of Physiology, Qingdao University Medical College, Qingdao, China.
| | | | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
4
|
Early-life Pb exposure as a potential risk factor for Alzheimer’s disease: are there hazards for the Mexican population? J Biol Inorg Chem 2019; 24:1285-1303. [DOI: 10.1007/s00775-019-01739-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/25/2019] [Indexed: 12/30/2022]
|
5
|
Li W, Su D, Zhai Q, Chi H, She X, Gao X, Wang K, Yang H, Wang R, Cui B. Proteomes analysis reveals the involvement of autophagy in AD-like neuropathology induced by noise exposure and ApoE4. ENVIRONMENTAL RESEARCH 2019; 176:108537. [PMID: 31228807 DOI: 10.1016/j.envres.2019.108537] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Noise is one of the most important environmental health hazards for humans. Environmental noise or apolipoprotein ε4 (ApoE4) can cause typical Alzheimer's disease (AD)-like pathological changes, which is characterized by progressive cognitive decline and neurodegenerative lesions. Gene-environment interactions may accelerate cognitive decline and increase AD risk. However, there is limited experimental evidence regarding the underlying mechanisms of noise-ApoE4 interactions and AD, which may be closely related to AD development. METHODS In this study, we investigated the combined effects of chronic noise exposure and the ApoE4 gene activation on hippocampus by using proteomics and differentially expressed proteins were found through performed gene ontology function and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. In addition, we assessed the changes in adult hippocampal neurogenesis and potential underlying mechanism for AD-like neuropathology. RESULTS Relative to control rats, combined exposure of noise and ApoE4 synergistically increased the characteristic pathological amyloid β-protein of AD-like neuropathology changes in hippocampus. The research identifies a total of 4147 proteins and 15 differentially expressed proteins in hippocampus. Furthermore, comparison of several of the diverse key pathways studied (e.g., PI3K/AKT, insulin, calpain-CDK5, and mammalian target of rapamycin (mTOR) signaling pathways) help to articulate the different mechanisms involved in combined effects of noise and ApoE4 on AD-like pathology. We verified four selected proteins, namely, eukaryotic translation elongation factor 1 epsilon 1, glycine amidinotransferase, nucleoredoxin, and tuberous sclerosis 1 proteins. Validation data shows significant effects of chronic noise and ApoE4 on the expression of four selected proteins, eukaryotic translation elongation factor 1 epsilon 1, glycine amidinotransferase, nucleoredoxin, and tuberous sclerosis 1 proteins, and mTOR and autophagy-related proteins, which share significant interaction effect of chronic noise and ApoE4. CONCLUSION Gene-environment interactions between chronic noise and ApoE4 activate the mTOR signaling, decrease autophagy, and facilitate AD-like changes in the hippocampus. Thus, our findings may help elucidate the role of gene-environment interactions in AD development.
Collapse
Affiliation(s)
- Wenlong Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China; School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Donghong Su
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Qingfeng Zhai
- School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Huimin Chi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China; School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Xiaojun She
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiujie Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Kun Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Honglian Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Rui Wang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Bo Cui
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.
| |
Collapse
|
6
|
Dou JF, Farooqui Z, Faulk CD, Barks AK, Jones T, Dolinoy DC, Bakulski KM. Perinatal Lead (Pb) Exposure and Cortical Neuron-Specific DNA Methylation in Male Mice. Genes (Basel) 2019; 10:genes10040274. [PMID: 30987383 PMCID: PMC6523909 DOI: 10.3390/genes10040274] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/25/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022] Open
Abstract
: Lead (Pb) exposure is associated with a wide range of neurological deficits. Environmental exposures may impact epigenetic changes, such as DNA methylation, and can affect neurodevelopmental outcomes over the life-course. Mating mice were obtained from a genetically invariant C57BL/6J background agouti viable yellow Avy strain. Virgin dams (a/a) were randomly assigned 0 ppm (control), 2.1 ppm (low), or 32 ppm (high) Pb-acetate water two weeks prior to mating with male mice (Avy/a), and this continued through weaning. At age 10 months, cortex neuronal nuclei were separated with NeuN⁺ antibodies in male mice to investigate neuron-specific genome-wide promoter DNA methylation using the Roche NimbleGen Mouse 3x720K CpG Island Promoter Array in nine pooled samples (three per dose). Several probes reached p-value < 10-5 , all of which were hypomethylated: 12 for high Pb (minimum false discovery rate (FDR) = 0.16, largest intensity ratio difference = -2.1) and 7 for low Pb (minimum FDR = 0.56, largest intensity ratio difference = -2.2). Consistent with previous results in bulk tissue, we observed a weak association between early-life exposure to Pb and DNA hypomethylation, with some affected genes related to neurodevelopment or cognitive function. Although these analyses were limited to males, data indicate that non-dividing cells such as neurons can be carriers of long-term epigenetic changes induced in development.
Collapse
Affiliation(s)
- John F Dou
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Zishaan Farooqui
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Christopher D Faulk
- Department of Animal Science, College of Food, Agricultural, and Natural Resource Sciences, University of Minnesota, St. Paul, MN 55108, USA.
| | - Amanda K Barks
- Department of Pediatrics, University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55454, USA.
| | - Tamara Jones
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Dana C Dolinoy
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Kelly M Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
7
|
Fathabadi B, Dehghanifiroozabadi M, Aaseth J, Sharifzadeh G, Nakhaee S, Rajabpour-Sanati A, Amirabadizadeh A, Mehrpour O. Comparison of Blood Lead Levels in Patients With Alzheimer's Disease and Healthy People. Am J Alzheimers Dis Other Demen 2018; 33:541-547. [PMID: 30134734 PMCID: PMC10852476 DOI: 10.1177/1533317518794032] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND It is argued that breakdown of β-amyloid in the brain causes deposition of senescent plaques and therefore Alzheimer's disease (AD). One of the influential factors for increasing level of this protein is exposure to lead. Our aim was to compare blood lead levels (BLLs) between patients with AD and healthy controls. METHODS This case-control study was performed on all patients with cognitive impairment who were referred to the Neurological Clinic of Birjand in 2016 to 2017. Patients were referred to the laboratory for measurement of their serum levels of lead. The controls and patients were matched by age and sex. RESULTS In the AD case group, the average BLL was 22.22 ± 28.57 μg/dL. Mann-Whitney U test showed that BLLs were significantly higher in the patients than in the controls. The unadjusted odds ratio for BLL among the patients was 1.05 (95% confidence interval: 1.01-1.09; P = .01) compared to the controls. CONCLUSION In the present study, BLL was associated with AD.
Collapse
Affiliation(s)
- Babak Fathabadi
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Dehghanifiroozabadi
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Department of Neurology, Birjand University of Medical Sciences, Complementary Alternative Medicine Research Center, Valiasr Hospital, Birjand, Iran
| | - Jan Aaseth
- Innlandet Hospital and Inland Norway University of Applied Sciences, Elverum, Norway
| | - Gholamreza Sharifzadeh
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Samaneh Nakhaee
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | - Ali Rajabpour-Sanati
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | - Alireza Amirabadizadeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
| | - Omid Mehrpour
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand, Iran
- Rocky Mountain Poison and Drug Center, Denver, CO, USA
| |
Collapse
|
8
|
Mansouri MT, Muñoz-Fambuena I, Cauli O. Cognitive impairment associated with chronic lead exposure in adults. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.npbr.2018.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
9
|
Hunter S, Smailagic N, Brayne C. Dementia Research: Populations, Progress, Problems, and Predictions. J Alzheimers Dis 2018; 64:S119-S143. [DOI: 10.3233/jad-179927] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sally Hunter
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Nadja Smailagic
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Cavallaro RA, Nicolia V, Fiorenza MT, Scarpa S, Fuso A. S-Adenosylmethionine and Superoxide Dismutase 1 Synergistically Counteract Alzheimer's Disease Features Progression in TgCRND8 Mice. Antioxidants (Basel) 2017; 6:antiox6040076. [PMID: 28973985 PMCID: PMC5745486 DOI: 10.3390/antiox6040076] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 01/30/2023] Open
Abstract
Recent evidence emphasizes the role of dysregulated one-carbon metabolism in Alzheimer’s Disease (AD). Exploiting a nutritional B-vitamin deficiency paradigm, we have previously shown that PSEN1 and BACE1 activity is modulated by one-carbon metabolism, leading to increased amyloid production. We have also demonstrated that S-adenosylmethionine (SAM) supplementation contrasted the AD-like features, induced by B-vitamin deficiency. In the present study, we expanded these observations by investigating the effects of SAM and SOD (Superoxide dismutase) association. TgCRND8 AD mice were fed either with a control or B-vitamin deficient diet, with or without oral supplementation of SAM + SOD. We measured oxidative stress by lipid peroxidation assay, PSEN1 and BACE1 expression by Real-Time Polymerase Chain Reaction (PCR), amyloid deposition by ELISA assays and immunohistochemistry. We found that SAM + SOD supplementation prevents the exacerbation of AD-like features induced by B vitamin deficiency, showing synergistic effects compared to either SAM or SOD alone. SAM + SOD supplementation also contrasts the amyloid deposition typically observed in TgCRND8 mice. Although the mechanisms underlying the beneficial effect of exogenous SOD remain to be elucidated, our findings identify that the combination of SAM + SOD could be carefully considered as co-adjuvant of current AD therapies.
Collapse
Affiliation(s)
- Rosaria A Cavallaro
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Vincenzina Nicolia
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Maria Teresa Fiorenza
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00183 Rome, Italy.
- IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64-65, 00143 Rome, Italy.
| | - Sigfrido Scarpa
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
| | - Andrea Fuso
- Department of Surgery "P. Valdoni", Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy.
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00183 Rome, Italy.
| |
Collapse
|
11
|
Bihaqi SW, Eid A, Zawia NH. Lead exposure and tau hyperphosphorylation: An in vitro study. Neurotoxicology 2017; 62:218-223. [DOI: 10.1016/j.neuro.2017.07.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/20/2017] [Accepted: 07/22/2017] [Indexed: 10/19/2022]
|
12
|
Engstrom AK, Snyder JM, Maeda N, Xia Z. Gene-environment interaction between lead and Apolipoprotein E4 causes cognitive behavior deficits in mice. Mol Neurodegener 2017; 12:14. [PMID: 28173832 PMCID: PMC5297175 DOI: 10.1186/s13024-017-0155-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/18/2017] [Indexed: 01/10/2023] Open
Abstract
Background Alzheimer’s disease (AD) is characterized by progressive cognitive decline and memory loss. Environmental factors and gene-environment interactions (GXE) may increase AD risk, accelerate cognitive decline, and impair learning and memory. However, there is currently little direct evidence supporting this hypothesis. Methods In this study, we assessed for a GXE between lead and ApoE4 on cognitive behavior using transgenic knock-in (KI) mice that express the human Apolipoprotein E4 allele (ApoE4-KI) or Apolipoprotein E3 allele (ApoE3-KI). We exposed 8-week-old male and female ApoE3-KI and ApoE4-KI mice to 0.2% lead acetate via drinking water for 12 weeks and assessed for cognitive behavior deficits during and after the lead exposure. In addition, we exposed a second (cellular) cohort of animals to lead and assessed for changes in adult hippocampal neurogenesis as a potential underlying mechanism for lead-induced learning and memory deficits. Results In the behavior cohort, we found that lead reduced contextual fear memory in all animals; however, this decrease was greatest and statistically significant only in lead-treated ApoE4-KI females. Similarly, only lead-treated ApoE4-KI females exhibited a significant decrease in spontaneous alternation in the T-maze. Furthermore, all lead-treated animals developed persistent spatial working memory deficits in the novel object location test, and this deficit manifested earlier in ApoE4-KI mice, with female ApoE4-KI mice exhibiting the earliest deficit onset. In the cellular cohort, we observed that the maturation, differentiation, and dendritic development of adult-born neurons in the hippocampus was selectively impaired in lead-treated female ApoE4-KI mice. Conclusions These data suggest that GXE between ApoE4 and lead exposure may contribute to cognitive impairment and that impaired adult hippocampal neurogenesis may contribute to these deficits in cognitive behavior. Together, these data suggest a role for GXE and sex differences in AD risk.
Collapse
Affiliation(s)
- Anna K Engstrom
- Toxicology Program, Department of Environmental and Occupational Health Sciences, University of Washington, Box 357234, Seattle, WA, 98195, USA
| | - Jessica M Snyder
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Nobuyo Maeda
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Zhengui Xia
- Toxicology Program, Department of Environmental and Occupational Health Sciences, University of Washington, Box 357234, Seattle, WA, 98195, USA.
| |
Collapse
|
13
|
Sanchez OF, Lee J, Yu King Hing N, Kim SE, Freeman JL, Yuan C. Lead (Pb) exposure reduces global DNA methylation level by non-competitive inhibition and alteration of dnmt expression. Metallomics 2017; 9:149-160. [DOI: 10.1039/c6mt00198j] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
14
|
Selwyn RG, Cooney SJ, Khayrullina G, Hockenbury N, Wilson CM, Jaiswal S, Bermudez S, Armstrong RC, Byrnes KR. Outcome after Repetitive Mild Traumatic Brain Injury Is Temporally Related to Glucose Uptake Profile at Time of Second Injury. J Neurotrauma 2016; 33:1479-91. [PMID: 26650903 DOI: 10.1089/neu.2015.4129] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Repeated mild traumatic brain injury (rmTBI) results in worsened outcomes, compared with a single injury, but the mechanism of this phenomenon is unclear. We have previously shown that mild TBI in a rat lateral fluid percussion model results in globally depressed glucose uptake, with a peak depression at 24 h that resolves by 16 days post-injury. The current study investigated the outcomes of a repeat injury conducted at various times during this period of depressed glucose uptake. Adult male rats were therefore subjected to rmTBI with a latency of 24 h, 5 days, or 15 days between injuries, followed by assessment of motor function, histopathology, and glucose uptake using positron emission tomography (PET). Rats that received a 24 h rmTBI showed significant deficits in motor function tasks, as well as significant increases in lesion volume and neuronal damage. The level of microglial and astrocytic activation also was associated with the timing of the second impact. Finally, rmTBI with latencies of 24 h and 5 days showed significant alterations in [(18)F]fluorodeoxyglucose uptake, compared with baseline scans. Therefore, we conclude that the state of the metabolic environment, as indicated by FDG-PET at the time of the repeat injury, significantly influences neurological outcomes.
Collapse
Affiliation(s)
- Reed G Selwyn
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
- 2 Department of Radiology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
- 3 Department of Radiology, University of New Mexico , Albuquerque, New Mexico
| | - Sean J Cooney
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
- 4 Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Guzal Khayrullina
- 4 Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Nicole Hockenbury
- 4 Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Colin M Wilson
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Shalini Jaiswal
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Sara Bermudez
- 4 Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Regina C Armstrong
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
- 4 Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Kimberly R Byrnes
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
- 4 Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| |
Collapse
|
15
|
Neurobehavioral impairments produced by developmental lead exposure persisted for generations in zebrafish (Danio rerio). Neurotoxicology 2016; 52:176-85. [DOI: 10.1016/j.neuro.2015.12.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/21/2015] [Accepted: 12/09/2015] [Indexed: 01/19/2023]
|
16
|
Szczygielski J, Mautes AE, Müller A, Sippl C, Glameanu C, Schwerdtfeger K, Steudel WI, Oertel J. Decompressive Craniectomy Increases Brain Lesion Volume and Exacerbates Functional Impairment in Closed Head Injury in Mice. J Neurotrauma 2015; 33:122-31. [PMID: 26102497 DOI: 10.1089/neu.2014.3835] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Decompressive craniectomy has been widely used in patients with head trauma. The randomized clinical trial on an early decompression (DECRA) demonstrated that craniectomy did not improve the neurological outcome, in contrast to previous animal experiments. The goal of our study was to analyze the effect of decompressive craniectomy in a murine model of head injury. Male mice were assigned into the following groups: sham, decompressive craniectomy, closed head injury (CHI), and CHI followed by craniectomy. At 24 h post-trauma, animals underwent the Neurological Severity Score test (NSS) and Beam Balance Score test (BBS). At the same time point, magnetic resonance imaging was performed, and volume of edema and contusion was assessed, followed by histopathological analysis. According to NSS, animals undergoing both trauma and craniectomy presented the most severe neurological impairment. Also, balancing time was reduced in this group compared with sham animals. Both edema and contusion volume were increased in the trauma and craniectomy group compared with sham animals. Histopathological analysis showed that all animals that underwent trauma presented substantial neuronal loss. In animals treated with craniectomy after trauma, a massive increase of edema with hemorrhagic transformation of contusion was documented. Decompressive craniectomy applied after closed head injury in mice leads to additional structural and functional impairment. The surgical decompression via craniectomy promotes brain edema formation and contusional blossoming in our model. This additive effect of combined mechanical and surgical trauma may explain the results of the DECRA trial and should be explored further in experiments.
Collapse
Affiliation(s)
- Jacek Szczygielski
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Angelika E Mautes
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Andreas Müller
- 2 Department of Radiology, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Christoph Sippl
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Cosmin Glameanu
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Karsten Schwerdtfeger
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Wolf-Ingo Steudel
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Joachim Oertel
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| |
Collapse
|
17
|
Fontana L, Rovina D, Novielli C, Maffioli E, Tedeschi G, Magnani I, Larizza L. Suggestive evidence on the involvement of polypyrimidine-tract binding protein in regulating alternative splicing of MAP/microtubule affinity-regulating kinase 4 in glioma. Cancer Lett 2015; 359:87-96. [PMID: 25578778 DOI: 10.1016/j.canlet.2014.12.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 12/23/2014] [Accepted: 12/26/2014] [Indexed: 01/01/2023]
Abstract
MAP/microtubule affinity-regulating kinase 4 (MARK4) is a serine-threonine kinase that phosphorylates microtubule-associated proteins taking part in the regulation of microtubule dynamics. MARK4 is expressed in two spliced isoforms characterized by inclusion (MARK4S) or exclusion (MARK4L) of exon 16. The distinct expression profiles in the central nervous system and their imbalance in gliomas point to roles of MARK4L and MARK4S in cell proliferation and cell differentiation, respectively. Having ruled out mutations and transcription defects, we hypothesized that alterations in the expression of splicing factors may underlie deregulated MARK4 expression in gliomas. Bioinformatic analysis revealed four putative polypyrimidine-tract binding (PTB) protein binding sites in MARK4 introns 15 and 16. Glioma tissues and glioblastoma-derived cancer stem cells showed, compared with normal brain, significant overexpression of PTB, correlated with high MARK4L mRNA expression. Splicing minigene assays revealed a functional intronic splicing silencer in MARK4 intron 15, but mutagenesis of the PTB binding site in this region did not affect minigene splicing, suggesting that PTB may bind to a splicing silencer other than the predicted one and synergistically acting with the other predicted PTB sites. Electrophoretic mobility shift assays coupled with mass spectrometry confirmed binding of PTB to the polypyrimidine tract of intron 15, and thus its involvement in MARK4 alternative splicing. This finding, along with evidence of PTB overexpression in gliomas and glioblastoma-derived cancer stem cells and differentiated progeny, merged in pointing out the involvement of PTB in the switch to MARK4L, consistent with its established role in driving oncogenic splicing in brain tumors.
Collapse
Affiliation(s)
- L Fontana
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - D Rovina
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - C Novielli
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - E Maffioli
- Department of Animal Pathology, Hygiene and Veterinary Public Health, Università degli Studi di Milano, Via Celoria, 10, 20133 Milan, Italy; Fondazione Filarete, Viale Ortles 22/4, 20139 Milan, Italy
| | - G Tedeschi
- Department of Animal Pathology, Hygiene and Veterinary Public Health, Università degli Studi di Milano, Via Celoria, 10, 20133 Milan, Italy; Fondazione Filarete, Viale Ortles 22/4, 20139 Milan, Italy
| | - I Magnani
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - L Larizza
- Department of Health Sciences, Medical Genetics, Università degli Studi di Milano, via Antonio di Rudinì 8, 20142 Milan, Italy; Laboratory of Medical Cytogenetics and Molecular Genetics, IRCCS Istituto Auxologico Italiano, Via Zucchi, 18, 20095 Cusano Milanino, Italy.
| |
Collapse
|
18
|
Hsieh CL, Niemi EC, Wang SH, Lee CC, Bingham D, Zhang J, Cozen ML, Charo I, Huang EJ, Liu J, Nakamura MC. CCR2 deficiency impairs macrophage infiltration and improves cognitive function after traumatic brain injury. J Neurotrauma 2014; 31:1677-88. [PMID: 24806994 DOI: 10.1089/neu.2013.3252] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Traumatic brain injury (TBI) provokes inflammatory responses, including a dramatic rise in brain macrophages in the area of injury. The pathway(s) responsible for macrophage infiltration of the traumatically injured brain and the effects of macrophages on functional outcomes are not well understood. C-C-chemokine receptor 2 (CCR2) is known for directing monocytes to inflamed tissues. To assess the role of macrophages and CCR2 in TBI, we determined outcomes in CCR2-deficient (Ccr2(-/-)) mice in a controlled cortical impact model. We quantified brain myeloid cell numbers post-TBI by flow cytometry and found that Ccr2(-/-) mice had greatly reduced macrophage numbers (∼80-90% reduction) early post-TBI, compared with wild-type mice. Motor, locomotor, and cognitive outcomes were assessed. Lack of Ccr2 improved locomotor activity with less hyperactivity in open field testing, but did not affect anxiety levels or motor coordination on the rotarod three weeks after TBI. Importantly, Ccr2(-/-) mice demonstrated greater spatial learning and memory, compared with wild-type mice eight weeks after TBI. Although there was no difference in the volume of tissue loss, Ccr2(-/-) mice had significantly increased neuronal density in the CA1-CA3 regions of the hippocampus after TBI, compared with wild-type mice. These data demonstrate that Ccr2 directs the majority of macrophage homing to the brain early after TBI and indicates that Ccr2 may facilitate harmful responses. Lack of Ccr2 improves functional recovery and neuronal survival. These results suggest that therapeutic blockade of CCR2-dependent responses may improve outcomes following TBI.
Collapse
Affiliation(s)
- Christine L Hsieh
- 1 Immunology Section, San Francisco VA Medical Center , San Francisco, California
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Navarro-Yepes J, Zavala-Flores L, Anandhan A, Wang F, Skotak M, Chandra N, Li M, Pappa A, Martinez-Fong D, Del Razo LM, Quintanilla-Vega B, Franco R. Antioxidant gene therapy against neuronal cell death. Pharmacol Ther 2014; 142:206-30. [PMID: 24333264 PMCID: PMC3959583 DOI: 10.1016/j.pharmthera.2013.12.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
Abstract
Oxidative stress is a common hallmark of neuronal cell death associated with neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, as well as brain stroke/ischemia and traumatic brain injury. Increased accumulation of reactive species of both oxygen (ROS) and nitrogen (RNS) has been implicated in mitochondrial dysfunction, energy impairment, alterations in metal homeostasis and accumulation of aggregated proteins observed in neurodegenerative disorders, which lead to the activation/modulation of cell death mechanisms that include apoptotic, necrotic and autophagic pathways. Thus, the design of novel antioxidant strategies to selectively target oxidative stress and redox imbalance might represent important therapeutic approaches against neurological disorders. This work reviews the evidence demonstrating the ability of genetically encoded antioxidant systems to selectively counteract neuronal cell loss in neurodegenerative diseases and ischemic brain damage. Because gene therapy approaches to treat inherited and acquired disorders offer many unique advantages over conventional therapeutic approaches, we discussed basic research/clinical evidence and the potential of virus-mediated gene delivery techniques for antioxidant gene therapy.
Collapse
Affiliation(s)
- Juliana Navarro-Yepes
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; Department of Toxicology, CINVESTAV-IPN, Mexico City, Mexico
| | - Laura Zavala-Flores
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Annadurai Anandhan
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Fang Wang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Maciej Skotak
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Namas Chandra
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, Alexandroupolis, Greece
| | - Daniel Martinez-Fong
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City, Mexico
| | | | | | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| |
Collapse
|
20
|
Senut MC, Sen A, Cingolani P, Shaik A, Land SJ, Ruden DM. Lead exposure disrupts global DNA methylation in human embryonic stem cells and alters their neuronal differentiation. Toxicol Sci 2014; 139:142-61. [PMID: 24519525 DOI: 10.1093/toxsci/kfu028] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Exposure to lead (Pb) during childhood can result in learning disabilities and behavioral problems. Although described in animal models, whether Pb exposure also alters neuronal differentiation in the developing brains of exposed children is unknown. Here, we investigated the effects of physiologically relevant concentrations of Pb (from 0.4 to 1.9μM) on the capacity of human embryonic stem cells (hESCs) to progress to a neuronal fate. We found that neither acute nor chronic exposure to Pb prevented hESCs from generating neural progenitor cells (NPCs). NPCs derived from hESCs chronically exposed to 1.9μM Pb throughout the neural differentiation process generated 2.5 times more TUJ1-positive neurons than those derived from control hESCs. Pb exposure of hESCs during the stage of neural rosette formation resulted in a significant decrease in the expression levels of the neural marker genes PAX6 and MSI1. Furthermore, the resulting NPCs differentiated into neurons with shorter neurites and less branching than control neurons, as assessed by Sholl analysis. DNA methylation studies of control, acutely treated hESCs and NPCs derived from chronically exposed hESCs using the Illumina HumanMethylation450 BeadChip demonstrated that Pb exposure induced changes in the methylation status of genes involved in neurogenetic signaling pathways. In summary, our study shows that exposure to Pb subtly alters the neuronal differentiation of exposed hESCs and that these changes could be partly mediated by modifications in the DNA methylation status of genes crucial to brain development.
Collapse
Affiliation(s)
- Marie-Claude Senut
- Institute of Environmental Health Sciences, C.S. Mott Center for Human Health and Development, Detroit, Michigan 48201
| | | | | | | | | | | |
Collapse
|
21
|
Blaney CE, Gunn RK, Stover KR, Brown RE. Maternal genotype influences behavioral development of 3×Tg-AD mouse pups. Behav Brain Res 2013; 252:40-8. [DOI: 10.1016/j.bbr.2013.05.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/15/2013] [Accepted: 05/20/2013] [Indexed: 12/19/2022]
|
22
|
Adwan L, Zawia NH. Epigenetics: a novel therapeutic approach for the treatment of Alzheimer's disease. Pharmacol Ther 2013; 139:41-50. [PMID: 23562602 PMCID: PMC3693222 DOI: 10.1016/j.pharmthera.2013.03.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 03/19/2013] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia in the elderly. It is characterized by the deposition of two forms of aggregates within the brain, the amyloid β plaques and tau neurofibrillary tangles. Currently, no disease-modifying agent is approved for the treatment of AD. Approved pharmacotherapies target the peripheral symptoms but they do not prevent or slow down the progression of the disease. Although several disease-modifying immunotherapeutic agents are in clinical development, many have failed due to the lack of efficacy or serious adverse events. Epigenetic changes including DNA methylation and histone modifications are involved in learning and memory and have been recently highlighted for holding promise as potential targets for AD therapeutics. Dynamic and latent epigenetic alterations are incorporated in AD pathological pathways and present valuable reversible targets for AD and other neurological disorders. The approval of epigenetic drugs for cancer treatment has opened the door for the development of epigenetic drugs for other disorders including neurodegenerative diseases. In particular, methyl donors and histone deacetylase inhibitors are being investigated for possible therapeutic effects to rescue memory and cognitive decline found in such disorders. This review explores the area of epigenetics for potential AD interventions and presents the most recent findings in this field.
Collapse
Affiliation(s)
- Lina Adwan
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Nasser H. Zawia
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
23
|
Lahiri DK, Sokol DK, Erickson C, Ray B, Ho CY, Maloney B. Autism as early neurodevelopmental disorder: evidence for an sAPPα-mediated anabolic pathway. Front Cell Neurosci 2013; 7:94. [PMID: 23801940 PMCID: PMC3689023 DOI: 10.3389/fncel.2013.00094] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 05/27/2013] [Indexed: 12/27/2022] Open
Abstract
Autism is a neurodevelopmental disorder marked by social skills and communication deficits and interfering repetitive behavior. Intellectual disability often accompanies autism. In addition to behavioral deficits, autism is characterized by neuropathology and brain overgrowth. Increased intracranial volume often accompanies this brain growth. We have found that the Alzheimer's disease (AD) associated amyloid-β precursor protein (APP), especially its neuroprotective processing product, secreted APP α, is elevated in persons with autism. This has led to the "anabolic hypothesis" of autism etiology, in which neuronal overgrowth in the brain results in interneuronal misconnections that may underlie multiple autism symptoms. We review the contribution of research in brain volume and of APP to the anabolic hypothesis, and relate APP to other proteins and pathways that have already been directly associated with autism, such as fragile X mental retardation protein, Ras small GTPase/extracellular signal-regulated kinase, and phosphoinositide 3 kinase/protein kinase B/mammalian target of rapamycin. We also present additional evidence of magnetic resonance imaging intracranial measurements in favor of the anabolic hypothesis. Finally, since it appears that APP's involvement in autism is part of a multi-partner network, we extend this concept into the inherently interactive realm of epigenetics. We speculate that the underlying molecular abnormalities that influence APP's contribution to autism are epigenetic markers overlaid onto potentially vulnerable gene sequences due to environmental influence.
Collapse
Affiliation(s)
- Debomoy K. Lahiri
- Department of Psychiatry, Indiana University School of MedicineIndianapolis, IN USA
- Laboratory of Medical and Molecular Genetics, Indiana University School of MedicineIndianapolis, IN, USA
- Institute of Psychiatric Research, Indiana University School of MedicineIndianapolis, IN, USA
| | - Deborah K. Sokol
- Department of Neurology, Indiana University School of MedicineIndianapolis, IN, USA
| | - Craig Erickson
- Cincinnati Children’s Hospital Medical CenterCincinnati, OH, USA
| | - Balmiki Ray
- Department of Psychiatry, Indiana University School of MedicineIndianapolis, IN USA
- Institute of Psychiatric Research, Indiana University School of MedicineIndianapolis, IN, USA
| | - Chang Y. Ho
- Department of Radiology and Imaging Sciences, Indiana University School of MedicineIndianapolis, IN, USA
| | - Bryan Maloney
- Department of Psychiatry, Indiana University School of MedicineIndianapolis, IN USA
- Institute of Psychiatric Research, Indiana University School of MedicineIndianapolis, IN, USA
| |
Collapse
|
24
|
Prenatal Exposures to Environmental Chemicals and Children's Neurodevelopment: An Update. Saf Health Work 2013; 4:1-11. [PMID: 23515885 PMCID: PMC3601292 DOI: 10.5491/shaw.2013.4.1.1] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 02/06/2013] [Accepted: 02/06/2013] [Indexed: 01/17/2023] Open
Abstract
This review surveys the recent literature on the neurodevelopmental impacts of chemical exposures during pregnancy. The review focuses primarily on chemicals of recent concern, including phthalates, bisphenol-A, polybrominated diphenyl ethers, and perfluorinated compounds, but also addresses chemicals with longer histories of investigation, including air pollutants, lead, methylmercury, manganese, arsenic, and organophosphate pesticides. For some chemicals of more recent concern, the available literature does not yet afford strong conclusions about neurodevelopment toxicity. In such cases, points of disagreement among studies are identified and suggestions provided for approaches to resolution of the inconsistencies, including greater standardization of methods for expressing exposure and assessing outcomes.
Collapse
|
25
|
De Coster S, van Larebeke N. Endocrine-disrupting chemicals: associated disorders and mechanisms of action. JOURNAL OF ENVIRONMENTAL AND PUBLIC HEALTH 2012; 2012:713696. [PMID: 22991565 PMCID: PMC3443608 DOI: 10.1155/2012/713696] [Citation(s) in RCA: 342] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 05/10/2012] [Accepted: 05/10/2012] [Indexed: 12/21/2022]
Abstract
The incidence and/or prevalence of health problems associated with endocrine-disruption have increased. Many chemicals have endocrine-disrupting properties, including bisphenol A, some organochlorines, polybrominated flame retardants, perfluorinated substances, alkylphenols, phthalates, pesticides, polycyclic aromatic hydrocarbons, alkylphenols, solvents, and some household products including some cleaning products, air fresheners, hair dyes, cosmetics, and sunscreens. Even some metals were shown to have endocrine-disrupting properties. Many observations suggesting that endocrine disruptors do contribute to cancer, diabetes, obesity, the metabolic syndrome, and infertility are listed in this paper. An overview is presented of mechanisms contributing to endocrine disruption. Endocrine disruptors can act through classical nuclear receptors, but also through estrogen-related receptors, membrane-bound estrogen-receptors, and interaction with targets in the cytosol resulting in activation of the Src/Ras/Erk pathway or modulation of nitric oxide. In addition, changes in metabolism of endogenous hormones, cross-talk between genomic and nongenomic pathways, cross talk with estrogen receptors after binding on other receptors, interference with feedback regulation and neuroendocrine cells, changes in DNA methylation or histone modifications, and genomic instability by interference with the spindle figure can play a role. Also it was found that effects of receptor activation can differ in function of the ligand.
Collapse
Affiliation(s)
| | - Nicolas van Larebeke
- Study Centre for Carcinogenesis and Primary Prevention of Cancer, Department of Radiotherapy and Experimental Cancerology, Ghent University Hospital, De Pintelaan 185 3K3, 9000 Ghent, Belgium
| |
Collapse
|
26
|
Affiliation(s)
- Kasper P Kepp
- DTU Chemistry, Technical University of Denmark, DK 2800 Kongens Lyngby, Denmark.
| |
Collapse
|
27
|
Balazs R, Vernon J, Hardy J. Epigenetic mechanisms in Alzheimer's disease: progress but much to do. Neurobiol Aging 2012; 32:1181-7. [PMID: 21669333 DOI: 10.1016/j.neurobiolaging.2011.02.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 02/02/2011] [Indexed: 12/22/2022]
Abstract
The interesting review from Mastroeni and colleagues highlights recent progress on epigenetic analysis of Alzheimer's disease, but it also illustrates how much we still need to do.
Collapse
Affiliation(s)
- Robert Balazs
- Reta Lilla Weston Research Laboratories and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK.
| | | | | |
Collapse
|
28
|
The role of clusterin in Alzheimer's disease: pathways, pathogenesis, and therapy. Mol Neurobiol 2012; 45:314-26. [PMID: 22274961 DOI: 10.1007/s12035-012-8237-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 01/12/2012] [Indexed: 10/14/2022]
Abstract
Genetic variation in clusterin gene, also known as apolipoprotein J, has been associated with Alzheimer's disease (AD) through replicated genome-wide studies, and plasma clusterin levels are associated with brain atrophy, baseline prevalence and severity, and rapid clinical progression in patients with AD, highlighting the importance of clusterin in AD pathogenesis. Emerging data suggest that clusterin contributes to AD through various pathways, including amyloid-β aggregation and clearance, lipid metabolism, neuroinflammation, and neuronal cell cycle control and apoptosis. Moreover, epigenetic regulation of the clusterin expression also seems to play an important role in the pathogenesis of AD. Emerging knowledge of the contribution of clusterin to the pathogenesis of AD presents new opportunities for AD therapy.
Collapse
|
29
|
Yang DJ, Shi S, Yao TM, Ji LN. The Impacts of Hg(II) Tightly Binding on the Alzheimer’s Tau Construct R3: Misfolding and Aggregation. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2011. [DOI: 10.1246/bcsj.20110133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
30
|
Valluru M, Staton CA, Reed MWR, Brown NJ. Transforming Growth Factor-β and Endoglin Signaling Orchestrate Wound Healing. Front Physiol 2011; 2:89. [PMID: 22164144 PMCID: PMC3230065 DOI: 10.3389/fphys.2011.00089] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 11/10/2011] [Indexed: 12/17/2022] Open
Abstract
Physiological wound healing is a complex process requiring the temporal and spatial co-ordination of various signaling networks, biomechanical forces, and biochemical signaling pathways in both hypoxic and non-hypoxic conditions. Although a plethora of factors are required for successful physiological tissue repair, transforming growth factor beta (TGF-β) expression has been demonstrated throughout wound healing and shown to regulate many processes involved in tissue repair, including production of ECM, proteases, protease inhibitors, migration, chemotaxis, and proliferation of macrophages, fibroblasts of the granulation tissue, epithelial and capillary endothelial cells. TGF-β mediates these effects by stimulating signaling pathways through a receptor complex which contains Endoglin. Endoglin is expressed in a broad spectrum of proliferating and stem cells with elevated expression during hypoxia, and regulates important cellular functions such as proliferation and adhesion via Smad signaling. This review focuses on how the TGF-β family and Endoglin, regulate stem cell availability, and modulate cellular behavior within the wound microenvironment, includes current knowledge of the signaling pathways involved, and explores how this information may be applicable to inflammatory and/or angiogenic diseases such as fibrosis, rheumatoid arthritis and metastatic cancer.
Collapse
Affiliation(s)
- Manoj Valluru
- Department of Oncology, Microcirculation Research Group, Faculty of Medicine, Dentistry and Health, University of Sheffield Sheffield, UK
| | | | | | | |
Collapse
|
31
|
Cooperative folding of tau peptide by coordination of group IIB metal cations during heparin-induced aggregation. Biometals 2011; 25:361-72. [DOI: 10.1007/s10534-011-9505-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 11/01/2011] [Indexed: 12/29/2022]
|
32
|
Manolopoulos VG, Ragia G, Tavridou A. Pharmacogenomics of oral antidiabetic medications: current data and pharmacoepigenomic perspective. Pharmacogenomics 2011; 12:1161-91. [PMID: 21843065 DOI: 10.2217/pgs.11.65] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is an increasingly prevalent disease. Several classes of drugs are currently available to treat T2DM patients; however, clinical response to these drugs often exhibits significant variation among individuals. For the oral antidiabetic drug classes of sulfonylureas, nonsulfonylurea insulin secretagogs, biguanides and thiazolidinediones, pharmacogenomic evidence has accumulated demonstrating an association between specific gene polymorphisms and interindividual variability in their therapeutic and adverse reaction effects. These polymorphisms are in genes of molecules involved in metabolism, transport and therapeutic mechanisms of the aforementioned drugs. Overall, it appears that pharmacogenomics has the potential to improve the management of T2DM and help clinicians in the effective prescribing of oral antidiabetic medications. Although pharmacogenomics can explain some of the heterogeneity in dose requirements, response and incidence of adverse effects of drugs between individuals, it is now clearly understood that much of the diversity in drug effects cannot be solely explained by studying the genomic diversity. Epigenomics, the field that focuses on nongenomic modifications that influence gene expression, may expand the scope of pharmacogenomics towards optimization of drug therapy. Therefore, pharmacoepigenomics, the combined analysis of genetic variations and epigenetic modifications, holds promise for the realization of personalized medicine. Although pharmacoepigenomics has so far been evaluated mainly in cancer pharmacotherapy, studies on epigenomic modifications during T2DM development provide useful data on the potential of pharmacoepigenomics to elucidate the mechanisms underlying interindividual response to oral antidiabetic treatment. In summary, the present article focuses on available data from pharmacogenomic studies of oral antidiabetic drugs and also provides an overview of T2DM epigenomic research, which has the potential to boost the development of pharmacoepigenomics in antidiabetic treatment.
Collapse
Affiliation(s)
- Vangelis G Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Dragana Campus, 68100 Alexandroupolis, Greece.
| | | | | |
Collapse
|
33
|
Schwartz J, Bellinger D, Glass T. Exploring potential sources of differential vulnerability and susceptibility in risk from environmental hazards to expand the scope of risk assessment. Am J Public Health 2011; 101 Suppl 1:S94-101. [PMID: 22021315 DOI: 10.2105/ajph.2011.300272] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Genetic factors, other exposures, individual disease states and allostatic load, psychosocial stress, and socioeconomic position all have the potential to modify the response to environmental exposures. Moreover, many of these modifiers covary with the exposure, leading to much higher risks in some subgroups. These are not theoretical concerns; rather, all these patterns have already been demonstrated in studies of the effects of lead and air pollution. However, recent regulatory impact assessments for these exposures have generally not incorporated these findings. Therefore, differential risk and vulnerability is a critically important but neglected area within risk assessment, and should be incorporated in the future.
Collapse
Affiliation(s)
- Joel Schwartz
- Department of Environmental Health and Epidemiology, Harvard School of Public Health, and Harvard Center for Risk Analysis, Harvard University, Boston, MA 02215, USA.
| | | | | |
Collapse
|
34
|
Zhu HL, Meng SR, Fan JB, Chen J, Liang Y. Fibrillization of human tau is accelerated by exposure to lead via interaction with His-330 and His-362. PLoS One 2011; 6:e25020. [PMID: 21966400 PMCID: PMC3180286 DOI: 10.1371/journal.pone.0025020] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 08/23/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Neurofibrillary tangles, mainly consisted of bundles of filaments formed by the microtubule-associated protein Tau, are a hallmark of Alzheimer disease. Lead is a potent neurotoxin for human being especially for the developing children, and Pb(2+) at high concentrations is found in the brains of patients with Alzheimer disease. However, it has not been reported so far whether Pb(2+) plays a role in the pathology of Alzheimer disease through interaction with human Tau protein and thereby mediates Tau filament formation. In this study, we have investigated the effect of Pb(2+) on fibril formation of recombinant human Tau fragment Tau(244-372) and its mutants at physiological pH. METHODOLOGY/PRINCIPAL FINDINGS As revealed by thioflavin T and 8-anilino-1-naphthalene sulfonic acid fluorescence, the addition of 5-40 µM Pb(2+) significantly accelerates the exposure of hydrophobic region and filament formation of wild-type Tau(244-372) on the investigated time scale. As evidenced by circular dichroism and Fourier transform infrared spectroscopy, fibrils formed by wild-type Tau(244-372) in the presence of 5-40 µM Pb(2+) contain more β-sheet structure than the same amount of fibrils formed by the protein in the absence of Pb(2+). However, unlike wild-type Tau(244-372), the presence of 5-40 µM Pb(2+) has no obvious effects on fibrillization kinetics of single mutants H330A and H362A and double mutant H330A/H362A, and fibrils formed by such mutants in the absence and in the presence of Pb(2+) contain similar amounts of β-sheet structure. The results from isothermal titration calorimetry show that one Pb(2+) binds to one Tau monomer via interaction with His-330 and His-362, with sub-micromolar affinity. CONCLUSIONS/SIGNIFICANCE We demonstrate for the first time that the fibrillization of human Tau protein is accelerated by exposure to lead via interaction with His-330 and His-362. Our results suggest the possible involvement of Pb(2+) in the pathogenesis of Alzheimer disease and provide critical insights into the mechanism of lead toxicity.
Collapse
Affiliation(s)
- Hai-Li Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Sheng-Rong Meng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jun-Bao Fan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jie Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yi Liang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
- * E-mail:
| |
Collapse
|
35
|
Martin DP, Anantharam V, Jin H, Witte T, Houk R, Kanthasamy A, Kanthasamy AG. Infectious prion protein alters manganese transport and neurotoxicity in a cell culture model of prion disease. Neurotoxicology 2011; 32:554-62. [PMID: 21871919 DOI: 10.1016/j.neuro.2011.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 07/20/2011] [Accepted: 07/21/2011] [Indexed: 01/26/2023]
Abstract
Protein misfolding and aggregation are considered key features of many neurodegenerative diseases, but biochemical mechanisms underlying protein misfolding and the propagation of protein aggregates are not well understood. Prion disease is a classical neurodegenerative disorder resulting from the misfolding of endogenously expressed normal cellular prion protein (PrP(C)). Although the exact function of PrP(C) has not been fully elucidated, studies have suggested that it can function as a metal binding protein. Interestingly, increased brain manganese (Mn) levels have been reported in various prion diseases indicating divalent metals also may play a role in the disease process. Recently, we reported that PrP(C) protects against Mn-induced cytotoxicity in a neural cell culture model. To further understand the role of Mn in prion diseases, we examined Mn neurotoxicity in an infectious cell culture model of prion disease. Our results show CAD5 scrapie-infected cells were more resistant to Mn neurotoxicity as compared to uninfected cells (EC(50)=428.8 μM for CAD5 infected cells vs. 211.6 μM for uninfected cells). Additionally, treatment with 300 μM Mn in persistently infected CAD5 cells showed a reduction in mitochondrial impairment, caspase-3 activation, and DNA fragmentation when compared to uninfected cells. Scrapie-infected cells also showed significantly reduced Mn uptake as measured by inductively coupled plasma-mass spectrometry (ICP-MS), and altered expression of metal transporting proteins DMT1 and transferrin. Together, our data indicate that conversion of PrP to the pathogenic isoform enhances its ability to regulate Mn homeostasis, and suggest that understanding the interaction of metals with disease-specific proteins may provide further insight to protein aggregation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Dustin P Martin
- Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicity, Ames, IA 50011, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Fragou D, Fragou A, Kouidou S, Njau S, Kovatsi L. Epigenetic mechanisms in metal toxicity. Toxicol Mech Methods 2011; 21:343-52. [DOI: 10.3109/15376516.2011.557878] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
37
|
Epigenetic mechanisms in Alzheimer's disease. Neurobiol Aging 2011; 32:1161-80. [PMID: 21482442 DOI: 10.1016/j.neurobiolaging.2010.08.017] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 07/20/2010] [Accepted: 08/07/2010] [Indexed: 12/20/2022]
Abstract
Epigenetic modifications help orchestrate sweeping developmental, aging, and disease-causing changes in phenotype by altering transcriptional activity in multiple genes spanning multiple biologic pathways. Although previous epigenetic research has focused primarily on dividing cells, particularly in cancer, recent studies have shown rapid, dynamic, and persistent epigenetic modifications in neurons that have significant neuroendocrine, neurophysiologic, and neurodegenerative consequences. Here, we provide a review of the major mechanisms for epigenetic modification and how they are reportedly altered in aging and Alzheimer's disease (AD). Because of their reach across the genome, epigenetic mechanisms may provide a unique integrative framework for the pathologic diversity and complexity of AD.
Collapse
|
38
|
Chua SM, Ng BY, Lee TS. Genetic and Genomic Aspects of Alzheimer's Disease. PROCEEDINGS OF SINGAPORE HEALTHCARE 2010. [DOI: 10.1177/201010581001900408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease (AD) is the most common chronic neurodegenerative disease today, afflicting 35 million people worldwide. Age is the major risk factor. The heritability of AD is estimated to be around 60%. Less than 5% of AD cases are familial with early-onset of disease caused by specific gene mutations. Genetic studies over the past 2 decades have provided invaluable insights into this complex disease. Here we review AD from the latest genome-wide association studies (GWAS), and a brief review of the transcriptomics, proteomics, metabolomics and epigenetics. Ultimately, a system-wide approach is essential to integrating the diverse and complicated findings into a meaningful understanding of AD.
Collapse
Affiliation(s)
- Sze-Ming Chua
- Department of Psychiatry, Singapore General Hospital, Singapore
| | - Beng-Yeong Ng
- Department of Psychiatry, Singapore General Hospital, Singapore
| | - Tih-Shih Lee
- Department of Psychiatry, Singapore General Hospital, Singapore
- Duke University Medical School, Durham, North Carolina, USA
| |
Collapse
|
39
|
Devi L, Alldred MJ, Ginsberg SD, Ohno M. Sex- and brain region-specific acceleration of β-amyloidogenesis following behavioral stress in a mouse model of Alzheimer's disease. Mol Brain 2010; 3:34. [PMID: 21059265 PMCID: PMC2988063 DOI: 10.1186/1756-6606-3-34] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 11/08/2010] [Indexed: 12/05/2022] Open
Abstract
Background It is hypothesized that complex interactions between multiple environmental factors and genetic factors are implicated in sporadic Alzheimer's disease (AD); however, the underlying mechanisms are poorly understood. Importantly, recent evidence reveals that expression and activity levels of the β-site APP cleaving enzyme 1 (BACE1), which initiates amyloid-β (Aβ) production, are elevated in AD brains. In this study, we investigated a molecular mechanism by which sex and stress interactions may accelerate β-amyloidogenesis and contribute to sporadic AD. Results We applied 5-day restraint stress (6 h/day) to the male and female 5XFAD transgenic mouse model of AD at the pre-pathological stage of disease, which showed little amyloid deposition under non-stressed control conditions. Exposure to the relatively brief behavioral stress increased levels of neurotoxic Aβ42 peptides, the β-secretase-cleaved C-terminal fragment (C99) and plaque burden in the hippocampus of female 5XFAD mice but not in that of male 5XFAD mice. In contrast, significant changes in the parameters of β-amyloidosis were not observed in the cerebral cortex of stressed male or female 5XFAD mice. We found that this sex- and brain region-specific acceleration of β-amyloidosis was accounted for by elevations in BACE1 and APP levels in response to adverse stress. Furthermore, not only BACE1 mRNA but also phosphorylation of the translation initiation factor eIF2α (a proposed mediator of the post-transcriptional upregulation of BACE1) was elevated in the hippocampus of stressed female 5XFAD mice. Conclusions Our results suggest that the higher prevalence of sporadic AD in women may be attributable to the vulnerability of female brains (especially, the hippocampus) to stressful events, which alter APP processing to favor the β-amyloidogenesis through the transcriptional and translational upregulation of BACE1 combined with elevations in its substrate APP.
Collapse
Affiliation(s)
- Latha Devi
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962, USA
| | | | | | | |
Collapse
|
40
|
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are two common neurodegenerative diseases that result in the progressive damage or death of neurons. Environmental agents have the potential to damage the developing and mature nervous system, resulting in neurodegenerative diseases. Heritable changes in gene expression that do not involve coding sequence modifications are referred to as ‘epigenetic’. These modifications include DNA methylation and downstream modification of histones. Environmental factors, including heavy metals and dietary folate intake, perturb neurodegenerative genes by epigenetic means, leading to altered gene expression and late-onset neurodegenerative diseases. Research into the genetic control of DNA methylation indicates an allelic skewing in a significant proportion of genes. This phenomenon may determine how an individual’s genetic makeup can alter the effect an environmental factor has on their risk of developing neurodegeneration. Finally, preliminary evidence using cell culture and transgenic animal models suggests that whole classes of pan-epigenetic modifiers will have significant protective effects against common neurodegenerative diseases.
Collapse
Affiliation(s)
- John BJ Kwok
- Neuroscience Research Australia, Randwick, NSW, Australia and University of New South Wales, Kensington, NSW, Australia
| |
Collapse
|
41
|
Lead-induced accumulation of beta-amyloid in the choroid plexus: role of low density lipoprotein receptor protein-1 and protein kinase C. Neurotoxicology 2010; 31:524-32. [PMID: 20488202 DOI: 10.1016/j.neuro.2010.05.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 05/11/2010] [Accepted: 05/11/2010] [Indexed: 11/24/2022]
Abstract
The choroid plexus (CP), constituting the blood-cerebrospinal fluid barrier, has the capacity to remove beta-amyloid (Abeta) from the cerebrospinal fluid. Our previous work indicates that exposure to lead (Pb) results in Abeta accumulation in the CP by decreasing the expression of low density lipoprotein receptor protein-1 (LRP1), a protein involved in the transport and clearance of Abeta. The current study was designed to explore the relationship between Abeta accumulation, protein kinase C (PKC) activity, and LRP1 status in the CP following Pb exposure. Confocal microscopy revealed that LRP1 was primarily localized in the cytosol of the CP in control rats and migrated distinctly towards the apical surface and the microvilli following acute Pb exposure (27 mg Pb/kg, i.p., 24h). Co-immunostaining revealed a co-localization of both PKC-delta and LRP1 in the cytosol of control rats, with a distinct relocalization of both towards the apical membrane following Pb exposure. Preincubation of the tissues with PKC-delta inhibitor rottlerin (2 microM) prior to Pb exposure in vitro, resulted in abolishing the Pb-induced relocalization of LRP1 to the apical surface. Importantly, a significant elevation in intracellular Abeta levels (p<0.01) was observed in the cytosol of the CP following Pb exposure, which was abolished following preincubation with rottlerin. In addition, rottlerin caused a relocalization of Abeta from the cytosol to the nucleus in both Pb-treated and control CP tissues. Finally, co-immunoprecipitation studies revealed a strong protein-protein interaction between LRP1 and PKC-delta in the CP. These studies suggest that Pb exposure disrupts Abeta homeostasis at the CP, owing partly to a Pb-induced relocalization of LRP1 via PKC-delta.
Collapse
|
42
|
|
43
|
Kovatsi L, Leda K, Georgiou E, Elisavet G, Ioannou A, Antrea I, Haitoglou C, Costas H, Tzimagiorgis G, George T, Tsoukali H, Helen T, Kouidou S, Sofia K. p16 promoter methylation in Pb2+ -exposed individuals. Clin Toxicol (Phila) 2010; 48:124-8. [PMID: 20199129 DOI: 10.3109/15563650903567091] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND One of the principle symptoms of lead poisoning is the development of neurological disorders. Neuronal response is closely related to DNA methylation changes. Aim. In this study, we estimated p16 methylation in nine individuals exposed to lead using methylation-specific polymerase chain reaction followed by analysis of the methylated cytosine content of the product by thermal denaturation. RESULTS We found that, based on lead blood concentration, lead-exposed individuals were divided into two groups. Among highly exposed individuals (blood Pb(2+) concentration = 51-100 microg/dL), we observed complete CpG methylation, whereas for low Pb(2+) concentrations (blood Pb(2+) concentration = 6-11 microg/dL), we observed partial methylation. CONCLUSION Our results show that among lead-overexposed individuals, p16 methylation is frequent and extensive, and suggest that DNA methylation could be involved in the mechanism by which lead induces neurotoxicity.
Collapse
Affiliation(s)
- Leda Kovatsi
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Epigenetics is a rapidly growing field and holds great promise for a range of human diseases, including brain disorders such as Rett syndrome, anxiety and depressive disorders, schizophrenia, Alzheimer disease and Huntington disease. This review is concerned with the pharmacology of epigenetics to treat disorders of the epigenome whether induced developmentally or manifested/acquired later in life. In particular, we will focus on brain disorders and their treatment by drugs that modify the epigenome. While the use of DNA methyl transferase inhibitors and histone deacetylase inhibitors in in vitro and in vivo models have demonstrated improvements in disease-related deficits, clinical trials in humans have been less promising. We will address recent advances in our understanding of the complexity of the epigenome with its many molecular players, and discuss evidence for a compromised epigenome in the context of an ageing or diseased brain. We will also draw on examples of species differences that may exist between humans and model systems, emphasizing the need for more robust pre-clinical testing. Finally, we will discuss fundamental issues to be considered in study design when targeting the epigenome.
Collapse
Affiliation(s)
- Pritika Narayan
- Department of Pharmacology and the National Research Centre for Growth and Development, The University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
45
|
Savica R, Grossardt BR, Carlin JM, Icen M, Bower JH, Ahlskog JE, Maraganore DM, Steensma DP, Rocca WA. Anemia or low hemoglobin levels preceding Parkinson disease: a case-control study. Neurology 2009; 73:1381-7. [PMID: 19858460 DOI: 10.1212/wnl.0b013e3181bd80c1] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE It has been suggested that anemia may be a risk factor for dementia, for restless legs syndrome, and for Parkinson disease (PD). Thus, we investigated the association of anemia with the subsequent risk of PD using a case-control study design. METHODS We used the medical records-linkage system of the Rochester Epidemiology Project to identify 196 subjects who developed PD in Olmsted County, Minnesota, from 1976 through 1995. Each incident case was matched by age (+/-1 year) and sex to a general population control. We reviewed the complete medical records of cases and controls in the system to detect anemia defined using the World Health Organization criteria. RESULTS Anemia was more common in the history of cases than of controls (odds ratio 2.00, 95% confidence interval 1.31-3.06, p = 0.001). The association remained significant after adjustment for cigarette smoking, exposure to pesticides, or hysterectomy (in women). The association was not significantly different between men and women, or between PD patients with or without rest tremor. Analyses stratified by time of onset of anemia showed a greater association for anemia that started 20 to 29 years before the onset of PD. Hemoglobin levels were slightly but consistently lower in cases than in controls across all ages. CONCLUSIONS Our results support an association between anemia experienced early in life and the later development of Parkinson disease. The interpretation of this association remains uncertain.
Collapse
Affiliation(s)
- R Savica
- Department of Neurology, College of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Nuutinen T, Suuronen T, Kauppinen A, Salminen A. Clusterin: a forgotten player in Alzheimer's disease. ACTA ACUST UNITED AC 2009; 61:89-104. [PMID: 19651157 DOI: 10.1016/j.brainresrev.2009.05.007] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 05/13/2009] [Accepted: 05/14/2009] [Indexed: 11/16/2022]
Abstract
Clusterin, also known as apolipoprotein J, is a versatile chaperone molecule which contains several amphipathic and coiled-coil alpha-helices, typical characteristics of small heat shock proteins. In addition, clusterin has three large intrinsic disordered regions, so-called molten globule domains, which can stabilize stressed protein structures. Twenty years ago, it was demonstrated that the expression of clusterin was clearly increased in Alzheimer's disease (AD). Later it was observed that clusterin can bind amyloid-beta peptides and prevent their fibrillization. Clusterin is also involved in the clearance of amyloid-beta peptides and fibrils by binding to megalin receptors and enhancing their endocytosis within glial cells. Clusterin is a complement inhibitor and can suppress complement activation observed in AD. Clusterin is also present in lipoprotein particles and regulates cholesterol and lipid metabolism of brain which is disturbed in AD. Clusterin is a stress-induced chaperone which is normally secreted but in conditions of cellular stress, it can be transported to cytoplasm where it can bind to Bax protein and inhibit neuronal apoptosis. Clusterin can also bind to Smad2/3 proteins and potentiate the neuroprotective TGFbeta signaling. An alternative splicing can produce a variant isoform of clusterin which can be translocated to nuclei where it induces apoptosis. The role of nuclear clusterin in AD needs to be elucidated. We will review here the extensive literature linking clusterin to AD and examine the recent progress in clusterin research with the respect to AD pathology. Though clusterin can be viewed as a multipotent guardian of brain, it is unable to prevent the progressive neuropathology in chronic AD.
Collapse
Affiliation(s)
- Tapio Nuutinen
- Department of Neuroscience and Neurology, University of Kuopio, P.O. Box 1627, FIN-70211 Kuopio, Finland
| | | | | | | |
Collapse
|
47
|
Bellinger DC. Interpreting epidemiologic studies of developmental neurotoxicity: conceptual and analytic issues. Neurotoxicol Teratol 2009; 31:267-74. [PMID: 19595760 DOI: 10.1016/j.ntt.2009.06.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 06/20/2009] [Accepted: 06/25/2009] [Indexed: 11/25/2022]
Abstract
This paper discusses issues pertaining to the validity, precision, and interpretation of epidemiologic studies of neurotoxicity. With regard to validity, the critical issues pertain to the appropriate strategy for confounder adjustment, particularly when confounders are complex, multi-faceted constructs, and to the need for greater clarity and transparency in articulating the causal relationships implicit in the analytic approach applied. With regard to precision, the critical issue is a need to identify the contributors to the substantial variability observed in the effect estimates that describe dose-response and dose-effect relationships. In addition to methodological sources, such as imprecision in estimating dose at the critical organ site(s), true inter-individual differences in susceptibility to a neurotoxicant could also contribute to the variability. Variability might be reduced by taking full account of factors such as co-exposures or health co-morbidities, genetic polymorphisms, and the social ecology of exposure. With regard to interpretation, we need to do a better job as a field conveying to risk assessors and others the ecological significance of the types of performance deficits observed following neurotoxicant exposure, emphasizing the distinction between individual and population risk. A final issue discussed is the need to define standards for the conduct, analysis, and reporting of epidemiologic studies of neurotoxicity, similar to those developed for other fields.
Collapse
Affiliation(s)
- David C Bellinger
- Children's Hospital Boston, Harvard Medical School, Harvard School of Public Health, Farley Basement Box 127, Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Sinclair DA, Oberdoerffer P. The ageing epigenome: damaged beyond repair? Ageing Res Rev 2009; 8:189-98. [PMID: 19439199 DOI: 10.1016/j.arr.2009.04.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 04/30/2009] [Accepted: 04/30/2009] [Indexed: 12/28/2022]
Abstract
Of all the proposed causes of ageing, DNA damage remains a leading, though still debated theory. Unlike most other types of age-related cellular damage, which can hypothetically be reversed, mutations in DNA are permanent. Such errors result in the accumulation of changes to RNA and protein sequences with age, and are tightly linked to cellular senescence and overall organ dysfunction. Over the past few years, an additional, more global role has emerged for the contribution of DNA damage and genomic instability to the ageing process. We, and others have found that DNA damage and the concomitant repair process can induce genome-wide epigenetic changes, which may promote a variety of age-related transcriptional and functional changes. Here, we discuss the link between DNA damage, chromatin alterations and ageing, an interplay that explains how seemingly random DNA damage could manifest in predictable phenotypic changes that define ageing, changes that may ultimately be reversible.
Collapse
|
49
|
Unwind: chronic stress exacerbates the deficits of Alzheimer's disease. Biol Psychiatry 2009; 65:916-7. [PMID: 19433151 DOI: 10.1016/j.biopsych.2009.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Accepted: 04/02/2009] [Indexed: 11/21/2022]
|
50
|
van Wijngaarden E, Campbell JR, Cory-Slechta DA. Bone lead levels are associated with measures of memory impairment in older adults. Neurotoxicology 2009; 30:572-80. [PMID: 19477197 DOI: 10.1016/j.neuro.2009.05.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 04/21/2009] [Accepted: 05/08/2009] [Indexed: 01/08/2023]
Abstract
Accumulating evidence suggests a link between lead exposure and memory impairment but assessments based on predictive and validated measures are lacking. We conducted a pilot study of 47 healthy subjects 55-67 years of age to examine associations between bone lead levels and 4 tests sensitive to the natural history of Mild Cognitive Impairment (MCI) and Alzheimer's disease (AD). These include three subtests of the Cambridge Neuropsychological Test Automated Battery (delayed match-to-sample, paired associates learning and spatial recognition memory) and the Montreal Cognitive Assessment Test. Bone lead concentrations were measured at the mid-shaft of the tibia and the calcaneus with K X-ray fluorescence. Higher tibial and calcaneal bone lead values were significantly (p<0.05) associated with lower performance levels on delayed match-to-sample and paired associates learning in unadjusted analyses with Spearman rank correlation coefficients of about 0.4. Multiple linear regression analyses (i.e., least-squares means of cognitive test scores across tertiles of lead exposure) adjusted for age, education and smoking status continued to show an association of higher calcaneal lead levels with increasing memory impairments on delayed match-to-sample (p=0.07). As might be expected, additional adjustment for history of hypertension reduced the strength of this association (p=0.19). Given the demonstrated impact of lead exposure on hypertension and the vascular etiology of certain dementias, we speculate that hypertension could play a mediating role in the association between lead exposure and memory impairment.
Collapse
Affiliation(s)
- Edwin van Wijngaarden
- Department of Community and Preventive Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States.
| | | | | |
Collapse
|