1
|
Abstract
Redox proteomics is a field of proteomics that is concerned with the characterization of the oxidation state of proteins to gain information about their modulated structure, function, activity, and involvement in different physiological pathways. Oxidative modifications of proteins have been shown to be implicated in normal physiological processes of cells as well as in pathomechanisms leading to the development of cancer, diabetes, neurodegenerative diseases, and some rare hereditary metabolic diseases, like classic galactosemia. Reactive oxygen species generate a variety of reversible and irreversible modifications in amino acid residue side chains and within the protein backbone. These oxidative post-translational modifications (Ox-PTMs) can participate in the activation of signal transduction pathways and mediate the toxicity of harmful oxidants. Thus the application of advanced redox proteomics technologies is important for gaining insights into molecular mechanisms of diseases. Mass-spectrometry-based proteomics is one of the most powerful methods that can be used to give detailed qualitative and quantitative information on protein modifications and allows us to characterize redox proteomes associated with diseases. This Review illustrates the role and biological consequences of Ox-PTMs under basal and oxidative stress conditions by focusing on protein carbonylation and S-glutathionylation, two abundant modifications with an impact on cellular pathways that have been intensively studied during the past decade.
Collapse
Affiliation(s)
- Atef Mannaa
- Borg AlArab Higher Institute of Engineering and Technology , New Borg AlArab City , Alexandria , Egypt
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty , University of Cologne , Joseph-Stelzmann-Str. 52 , 50931 Cologne , Germany
| |
Collapse
|
2
|
Fernández MN, Muñoz-Olivas R, Luque-Garcia JL. SILAC-based quantitative proteomics identifies size-dependent molecular mechanisms involved in silver nanoparticles-induced toxicity. Nanotoxicology 2019; 13:812-826. [DOI: 10.1080/17435390.2019.1579374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- M. N. Fernández
- Faculty of Chemical Sciences, Department of Analytical Chemistry, Complutense University of Madrid, Madrid, Spain
| | - R. Muñoz-Olivas
- Faculty of Chemical Sciences, Department of Analytical Chemistry, Complutense University of Madrid, Madrid, Spain
| | - J. L. Luque-Garcia
- Faculty of Chemical Sciences, Department of Analytical Chemistry, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
3
|
Aldehyde dehydrogenase 2 in the spotlight: The link between mitochondria and neurodegeneration. Neurotoxicology 2018; 68:19-24. [PMID: 29936317 DOI: 10.1016/j.neuro.2018.06.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/08/2018] [Accepted: 06/11/2018] [Indexed: 01/18/2023]
Abstract
Growing body of evidence suggests that mitochondrial dysfunctions and resultant oxidative stress are likely responsible for many neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Aldehyde dehydrogenase (ALDH) superfamily plays a crucial role in several biological processes including development and detoxification pathways in the organism. In particular, ALDH2 is crucial in the oxidative metabolism of toxic aldehydes in the brain, such as catecholaminergic metabolites (DOPAL and DOPEGAL) and the principal product of lipid peroxidation process 4-HNE. This review aims to deepen the current knowledge regarding to ALDH2 function and its relation with brain-damaging processes that increase the risk to develop neurodegenerative disorders. We focused on relevant literature of what is currently known at molecular and cellular levels in experimental models of these pathologies. The understanding of ALDH2 contributions could be a potential target in new therapeutic approaches for PD and AD due to its crucial role in mitochondrial normal function maintenance that protects against neurotoxicity.
Collapse
|
4
|
Dammalli M, Dey G, Madugundu AK, Kumar M, Rodrigues B, Gowda H, Siddaiah BG, Mahadevan A, Shankar SK, Prasad TSK. Proteomic Analysis of the Human Olfactory Bulb. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:440-453. [PMID: 28816642 DOI: 10.1089/omi.2017.0084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The importance of olfaction to human health and disease is often underappreciated. Olfactory dysfunction has been reported in association with a host of common complex diseases, including neurological diseases such as Alzheimer's disease and Parkinson's disease. For health, olfaction or the sense of smell is also important for most mammals, for optimal engagement with their environment. Indeed, animals have developed sophisticated olfactory systems to detect and interpret the rich information presented to them to assist in day-to-day activities such as locating food sources, differentiating food from poisons, identifying mates, promoting reproduction, avoiding predators, and averting death. In this context, the olfactory bulb is a vital component of the olfactory system receiving sensory information from the axons of the olfactory receptor neurons located in the nasal cavity and the first place that processes the olfactory information. We report in this study original observations on the human olfactory bulb proteome in healthy subjects, using a high-resolution mass spectrometry-based proteomic approach. We identified 7750 nonredundant proteins from human olfactory bulbs. Bioinformatics analysis of these proteins showed their involvement in biological processes associated with signal transduction, metabolism, transport, and olfaction. These new observations provide a crucial baseline molecular profile of the human olfactory bulb proteome, and should assist the future discovery of biomarker proteins and novel diagnostics associated with diseases characterized by olfactory dysfunction.
Collapse
Affiliation(s)
- Manjunath Dammalli
- 1 Institute of Bioinformatics , Bangalore, India .,2 Department of Biotechnology, Siddaganga Institute of Technology , Tumakuru, India
| | - Gourav Dey
- 1 Institute of Bioinformatics , Bangalore, India .,3 Department of Biotechnology, Manipal University , Manipal, India
| | - Anil K Madugundu
- 1 Institute of Bioinformatics , Bangalore, India .,4 Centre for Bioinformatics, School of Life Sciences, Pondicherry University , Puducherry, India
| | - Manish Kumar
- 1 Institute of Bioinformatics , Bangalore, India .,3 Department of Biotechnology, Manipal University , Manipal, India
| | | | - Harsha Gowda
- 1 Institute of Bioinformatics , Bangalore, India .,5 YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University , Mangalore, India
| | | | - Anita Mahadevan
- 6 Department of Neuropathology, National Institute of Mental Health and Neurosciences , Bangalore, India .,7 Human Brain Tissue Repository, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences , Bangalore, India
| | - Susarla Krishna Shankar
- 6 Department of Neuropathology, National Institute of Mental Health and Neurosciences , Bangalore, India .,7 Human Brain Tissue Repository, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences , Bangalore, India .,8 NIMHANS-IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences , Bangalore, India
| | - Thottethodi Subrahmanya Keshava Prasad
- 1 Institute of Bioinformatics , Bangalore, India .,5 YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University , Mangalore, India .,8 NIMHANS-IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences , Bangalore, India
| |
Collapse
|
5
|
Yoo DY, Jung HY, Kim JW, Yim HS, Kim DW, Nam H, Suh JG, Choi JH, Won MH, Yoon YS, Hwang IK. Reduction of dynamin 1 in the hippocampus of aged mice is associated with the decline in hippocampal‑dependent memory. Mol Med Rep 2016; 14:4755-4760. [PMID: 27748822 DOI: 10.3892/mmr.2016.5804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 09/07/2016] [Indexed: 11/06/2022] Open
Abstract
Dynamin 1 is a known synaptic protein, which has is key in the presynaptic regulation of endocytosis. The present study investigated the association between age and the observed changes in Morris water maze performance, and immunoreactivity and protein levels of dynamin 1 in the mouse hippocampal formation. In addition, the effects of dynasore, an inhibitor of dynamin 1, on the hippocampal dependent memory were determined to elucidate the correlation between dynamin 1 and memory. In the training phase of the Morris water maze task, the mean escape latency of the aged group (24 months old) was significantly longer, compared with that of the adult group (4 months old), although the average swimming speed and the total distance traveled during the probe trial were similar in the two groups. In the aged group, the time spent locating the target platform was significantly longer and the time spent in the correct quadrant was significantly shorter, compared with those in the adult group. In the adult group, a moderate level of dynamin 1 was detected in the hippocampal CA1 and CA3 regions, and in the dentate gyrus. In the aged group, the immunoreactivity of dynamin 1 was almost eliminated in the CA3 region and the dentate gyrus. In addition, the protein levels of dynamin 1 in the brain were significantly lower in the aged group, compared with those in the adult group. The direct infusion of dynasore, significantly reduced the contextual memory, compared with that of animals in the vehicle‑treated group. These results suggested that dynamin 1 was susceptible to the aging process, and that a reduction in dynamin 1 may result in hippocampal‑dependent memory deficits by disrupting endocytosis and the release of neurotransmitters.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee Sun Yim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangneung‑Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangneung‑Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Hajin Nam
- Department of Medical Genetics, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Jun Gyo Suh
- Department of Medical Genetics, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
6
|
Lachén-Montes M, Fernández-Irigoyen J, Santamaría E. Deconstructing the molecular architecture of olfactory areas using proteomics. Proteomics Clin Appl 2016; 10:1178-1190. [PMID: 27226001 DOI: 10.1002/prca.201500147] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/02/2016] [Accepted: 05/22/2016] [Indexed: 11/07/2022]
Abstract
The anatomy of the olfactory system is highly complex, comprising a system of olfactory receptors, pathways for the transmission of olfactory information, and structures for the recognition, discrimination, and memorization of odors. During the last years, proteomics has emerged as a large-scale comprehensive approach to characterize and quantify specific olfactory-related proteomes in different biological conditions such as olfactory learning, neurodegeneration, and ageing between others. The current work reviews recent applications of proteomics to olfaction with particular focus on quantitative proteome profiling studies performed on olfactory areas from laboratory animal models as well as proteomic characterizations performed on specific brain structures and fluids involved in human smell. Finally, we will also discuss the potential application of proteomics to study global proteome dynamics and posttranslationally modified proteomes in order to unravel cell-signaling networks that occur from peripheral structures to olfactory cortical areas during odor processing.
Collapse
Affiliation(s)
- Mercedes Lachén-Montes
- Clinical Neuroproteomics Group, Navarrabiomed, Instituto de investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Group, Navarrabiomed, Instituto de investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Proteomics Unit, Navarrabiomed, Proteored-ISCIII, Instituto de investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Group, Navarrabiomed, Instituto de investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Proteomics Unit, Navarrabiomed, Proteored-ISCIII, Instituto de investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
7
|
A platelet protein biochip rapidly detects an Alzheimer's disease-specific phenotype. Acta Neuropathol 2014; 128:665-77. [PMID: 25248508 PMCID: PMC4201753 DOI: 10.1007/s00401-014-1341-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 10/25/2022]
Abstract
Alzheimer's disease (AD), a multifactorial neurodegenerative condition caused by genetic and environmental factors, is diagnosed using neuropsychological tests and brain imaging; molecular diagnostics are not routinely applied. Studies have identified AD-specific cerebrospinal fluid (CSF) biomarkers but sample collection requires invasive lumbar puncture. To identify AD-modulated proteins in easily accessible blood platelets, which share biochemical signatures with neurons, we compared platelet lysates from 62 AD, 24 amnestic mild cognitive impairment (aMCI), 13 vascular dementia (VaD), and 12 Parkinson's disease (PD) patients with those of 112 matched controls by fluorescence two-dimensional differential gel electrophoresis in independent discovery and verification sets. The optimal sum score of four mass spectrometry (MS)-identified proteins yielded a sensitivity of 94 % and a specificity of 89 % (AUC = 0.969, 95 % CI = 0.944-0.994) to differentiate AD patients from healthy controls. To bridge the gap between bench and bedside, we developed a high-throughput multiplex protein biochip with great potential for routine AD screening. For convenience and speed of application, this array combines loading control-assisted protein quantification of monoamine oxidase B and tropomyosin 1 with protein-based genotyping for single nucleotide polymorphisms (SNPs) in the apolipoprotein E and glutathione S-transferase omega 1 genes. Based on minimally invasive blood drawing, this innovative protein biochip enables identification of AD patients with an accuracy of 92 % in a single analytical step in less than 4 h.
Collapse
|
8
|
Butterfield DA, Gu L, Di Domenico F, Robinson RAS. Mass spectrometry and redox proteomics: applications in disease. MASS SPECTROMETRY REVIEWS 2014; 33:277-301. [PMID: 24930952 DOI: 10.1002/mas.21374] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/07/2013] [Accepted: 02/07/2013] [Indexed: 06/03/2023]
Abstract
Proteomics techniques are continuously being developed to further understanding of biology and disease. Many of the pathways that are relevant to disease mechanisms rely on the identification of post-translational modifications (PTMs) such as phosphorylation, acetylation, and glycosylation. Much attention has also been focused on oxidative PTMs which include protein carbonyls, protein nitration, and the incorporation of fatty acids and advanced glycation products to amino acid side chains, amongst others. The introduction of these PTMs in the cell can occur due to the attack of reactive oxygen and nitrogen species (ROS and RNS, respectively) on proteins. ROS and RNS can be present as a result of normal metabolic processes as well as external factors such as UV radiation, disease, and environmental toxins. The imbalance of ROS and RNS with antioxidant cellular defenses leads to a state of oxidative stress, which has been implicated in many diseases. Redox proteomics techniques have been used to characterize oxidative PTMs that result as a part of normal cell signaling processes as well as oxidative stress conditions. This review highlights many of the redox proteomics techniques which are currently available for several oxidative PTMs and brings to the reader's attention the application of redox proteomics for understanding disease pathogenesis in neurodegenerative disorders and others such as cancer, kidney, and heart diseases.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, 40506
| | | | | | | |
Collapse
|
9
|
Brann JH, Firestein SJ. A lifetime of neurogenesis in the olfactory system. Front Neurosci 2014; 8:182. [PMID: 25018692 PMCID: PMC4071289 DOI: 10.3389/fnins.2014.00182] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/09/2014] [Indexed: 12/11/2022] Open
Abstract
Neurogenesis continues well beyond embryonic and early postnatal ages in three areas of the nervous system. The subgranular zone supplies new neurons to the dentate gyrus of the hippocampus. The subventricular zone supplies new interneurons to the olfactory bulb, and the olfactory neuroepithelia generate new excitatory sensory neurons that send their axons to the olfactory bulb. The latter two areas are of particular interest as they contribute new neurons to both ends of a first-level circuit governing olfactory perception. The vomeronasal organ and the main olfactory epithelium comprise the primary peripheral olfactory epithelia. These anatomically distinct areas share common features, as each exhibits extensive neurogenesis well beyond the juvenile phase of development. Here we will discuss the effect of age on the structural and functional significance of neurogenesis in the vomeronasal and olfactory epithelia, from juvenile to advanced adult ages, in several common model systems. We will next discuss how age affects the regenerative capacity of these neural stem cells in response to injury. Finally, we will consider the integration of newborn neurons into an existing circuit as it is modified by the age of the animal.
Collapse
Affiliation(s)
- Jessica H Brann
- Department of Biology, Loyola University Chicago Chicago, IL, USA
| | - Stuart J Firestein
- Department of Biological Sciences, Columbia University New York, NY, USA ; Department of Neuroscience, Columbia University New York, NY, USA
| |
Collapse
|
10
|
Abstract
The scope of the current paper is to review existing and potential applications of proteomic analysis to aging research. The focus will lie on the unique opportunities of high-throughput studies for uncovering specific alterations in protein expression, protein complexes or protein modifications caused by biological aging. The result of such studies will outline aging phenotypes and potentially indicate pathways involved in the pathogenesis of age-associated disfunctions. Specific attention is paid to the illustrations of successful applications of proteomic technologies and potential applications of new proteomic concepts to biogerontological studies.
Collapse
Affiliation(s)
- Victor S Sharov
- University of Kansas, Pharmaceutical Chemistry Department, Lawrence, KS 66047, USA.
| | | |
Collapse
|
11
|
Redox proteomics and the dynamic molecular landscape of the aging brain. Ageing Res Rev 2014; 13:75-89. [PMID: 24374232 DOI: 10.1016/j.arr.2013.12.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 12/04/2013] [Accepted: 12/16/2013] [Indexed: 12/13/2022]
Abstract
It is well established that the risk to develop neurodegenerative disorders increases with chronological aging. Accumulating studies contributed to characterize the age-dependent changes either at gene and protein expression level which, taken together, show that aging of the human brain results from the combination of the normal decline of multiple biological functions with environmental factors that contribute to defining disease risk of late-life brain disorders. Finding the "way out" of the labyrinth of such complex molecular interactions may help to fill the gap between "normal" brain aging and development of age-dependent diseases. To this purpose, proteomics studies are a powerful tool to better understand where to set the boundary line of healthy aging and age-related disease by analyzing the variation of protein expression levels and the major post translational modifications that determine "protein" physio/pathological fate. Increasing attention has been focused on oxidative modifications due to the crucial role of oxidative stress in aging, in addition to the fact that this type of modification is irreversible and may alter protein function. Redox proteomics studies contributed to decipher the complexity of brain aging by identifying the proteins that were increasingly oxidized and eventually dysfunctional as a function of age. The purpose of this review is to summarize the most important findings obtained by applying proteomics approaches to murine models of aging with also a brief overview of some human studies, in particular those related to dementia.
Collapse
|
12
|
Fernández-Irigoyen J, Corrales FJ, Santamaría E. Proteomic atlas of the human olfactory bulb. J Proteomics 2012; 75:4005-4016. [PMID: 22609191 DOI: 10.1016/j.jprot.2012.05.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 05/04/2012] [Accepted: 05/07/2012] [Indexed: 11/28/2022]
Abstract
The olfactory bulb (OB) is the first site for the processing of olfactory information in the brain and its deregulation is associated with neurodegenerative disorders. Although different efforts have been made to characterize the human brain proteome in depth, the protein composition of the human OB remains largely unexplored. We have performed a comprehensive analysis of the human OB proteome employing protein and peptide fractionation methods followed by LC-MS/MS, identifying 1529 protein species, corresponding to 1466 unique proteins, which represents a 7-fold increase in proteome coverage with respect to previous OB proteome descriptions from translational models. Bioinformatic analyses revealed that protein components of the OB participated in a plethora of biological process highlighting hydrolase and phosphatase activities and nucleotide and RNA binding activities. Interestingly, 631 OB proteins identified were not previously described in protein datasets derived from large-scale Human Brain Proteome Project (HBPP) studies. In particular, a subset of these differential proteins was mainly involved in axon guidance, opioid signaling, neurotransmitter receptor binding, and synaptic plasticity. Taken together, these results increase our knowledge about the molecular composition of the human OB and may be useful to understand the molecular basis of the olfactory system and the etiology of its disorders.
Collapse
|
13
|
Robinson RAS, Joshi G, Huang Q, Sultana R, Baker AS, Cai J, Pierce W, St Clair DK, Markesbery WR, Butterfield DA. Proteomic analysis of brain proteins in APP/PS-1 human double mutant knock-in mice with increasing amyloid β-peptide deposition: insights into the effects of in vivo treatment with N-acetylcysteine as a potential therapeutic intervention in mild cognitive impairment and Alzheimer's disease. Proteomics 2011; 11:4243-56. [PMID: 21954051 DOI: 10.1002/pmic.201000523] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 07/21/2011] [Accepted: 08/18/2011] [Indexed: 12/14/2022]
Abstract
Proteomics analyses were performed on the brains of wild-type (WT) controls and an Alzheimer's disease (AD) mouse model, APP/PS-1 human double mutant knock-in mice. Mice were given either drinking water or water supplemented with N-acetylcysteine (NAC) (2 mg/kg body weight) for a period of five months. The time periods of treatment correspond to ages prior to Aβ deposition (i.e. 4-9 months), resembling human mild cognitive impairment (MCI), and after Aβ deposition (i.e. 7-12 months), more closely resembling advancing stages of AD. Substantial differences exist between the proteomes of WT and APP/PS-1 mice at 9 or 12 months, indicating that Aβ deposition and oxidative stress lead to downstream changes in protein expression. Altered proteins are involved in energy-related pathways, excitotoxicity, cell cycle signaling, synaptic abnormalities, and cellular defense and structure. Overall, the proteomic results support the notion that NAC may be beneficial for increasing cellular stress responses in WT mice and for influencing the levels of energy- and mitochondria-related proteins in APP/PS-1 mice.
Collapse
Affiliation(s)
- Renã A S Robinson
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Sultana R, Di Domenico F, Tseng M, Cai J, Noel T, Chelvarajan RL, Pierce WD, Cini C, Bondada S, St. Clair DK, Butterfield DA. Doxorubicin-Induced Thymus Senescence. J Proteome Res 2010; 9:6232-41. [DOI: 10.1021/pr100465m] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Rukhsana Sultana
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - Fabio Di Domenico
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - Michael Tseng
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - Jian Cai
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - Teresa Noel
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - R. Lakshman Chelvarajan
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - William D. Pierce
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - Ciara Cini
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - Subbarao Bondada
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - Daret K. St. Clair
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| | - D. Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy, Department of Anatomical Sciences & Neurobiology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Pharmacology, University of Louisville, Louisville, Kentucky 40202, United States, Department of Toxicology, University of Kentucky, Lexington, Kentucky 40536, United States, Department of Microbiology
| |
Collapse
|
15
|
Di Domenico F, Perluigi M, Butterfield DA, Cornelius C, Calabrese V. Oxidative damage in rat brain during aging: interplay between energy and metabolic key target proteins. Neurochem Res 2010; 35:2184-92. [PMID: 20963486 DOI: 10.1007/s11064-010-0295-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2010] [Indexed: 12/21/2022]
Abstract
Aging is characterized by a gradual and continuous loss of physiological functions and responses particularly marked in the central nervous system. Reactive oxygen species (ROS) can react with all major biological macromolecules such as carbohydrates, nucleic acids, lipids, and proteins. Since proteins are the major components of biological systems and regulate multiple cellular pathways, oxidative damage of key proteins are considered to be the principal molecular mechanisms leading to age-related deficits. Recent evidences support the notion that a decrease of energy metabolism in the brain contribute to neuronal loss and cognitive decline associated with aging. In the present study we identified selective protein targets which are oxidized in aged rats compared with adult rats. Most of the oxidatively modified proteins we found in the present study are key proteins involved in energy metabolism and ATP production. Oxidative modification of these proteins was associated with decreased enzyme activities. In addition, we also found decreased levels of thiol reducing system. Our study demonstrated that oxidative damage to specific proteins impairs energy metabolism and ATP production thus contributing to shift neuronal cells towards a more oxidized environment which ultimately might compromise multiple neuronal functions. These results further confirm that increased protein oxidation coupled with decreased reducing systems are characteristic hallmarks of aging and aging-related degenerative processes.
Collapse
Affiliation(s)
- F Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, P le A Moro 5, 00185 Rome, Italy
| | | | | | | | | |
Collapse
|
16
|
Perluigi M, Di Domenico F, Giorgi A, Schininà ME, Coccia R, Cini C, Bellia F, Cambria MT, Cornelius C, Butterfield DA, Calabrese V. Redox proteomics in aging rat brain: involvement of mitochondrial reduced glutathione status and mitochondrial protein oxidation in the aging process. J Neurosci Res 2010; 88:3498-507. [PMID: 20936692 DOI: 10.1002/jnr.22500] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2010] [Revised: 07/11/2010] [Accepted: 07/16/2010] [Indexed: 12/14/2022]
Abstract
Increasing evidence supports the notion that increased oxidative stress is a fundamental cause in the aging process and in neurodegenerative diseases. As a result, a decline in cognitive function is generally associated with brain aging. Reactive oxygen species (ROS) are highly reactive intermediates, which can modify proteins, nucleic acids, and polyunsaturated fatty acids, leading to neuronal damage. Because proteins are major components of biological systems and play key roles in a variety of cellular functions, oxidative damage to proteins represents a primary event observed in aging and age-related neurodegenerative disorders. In the present study, with a redox proteomics approach, we identified mitochondrial oxidatively modified proteins as a function of brain aging, specifically in those brain regions, such as cortex and hippocampus, that are commonly affected by the aging process. In all brain regions examined, many of the identified proteins were energy-related, such as pyruvate kinase, ATP synthase, aldolase, creatine kinase, and α-enolase. These alterations were associated with significant changes in both cytosolic and mitochondrial redox status in all brain regions analyzed. Our finding is in line with current literature postulating that free radical damage and decreased energy production are characteristic hallmarks of the aging process. In additon, our results further contribute to identifying common pathological pathways involved both in aging and in neurodegenerative disease development.
Collapse
Affiliation(s)
- M Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Joshi G, Aluise CD, Cole MP, Sultana R, Pierce WM, Vore M, St Clair DK, Butterfield DA. Alterations in brain antioxidant enzymes and redox proteomic identification of oxidized brain proteins induced by the anti-cancer drug adriamycin: implications for oxidative stress-mediated chemobrain. Neuroscience 2010; 166:796-807. [PMID: 20096337 DOI: 10.1016/j.neuroscience.2010.01.021] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 01/13/2010] [Accepted: 01/13/2010] [Indexed: 12/30/2022]
Abstract
Adriamycin (ADR) is a chemotherapeutic for the treatment of solid tumors. This quinone-containing anthracycline is well known to produce large amounts of reactive oxygen species (ROS) in vivo. A common complaint of patients undergoing long-term treatment with ADR is somnolence, often referred to as "chemobrain." While ADR itself does not cross the blood brain barrier (BBB), we recently showed that ADR administration causes a peripheral increase in tumor necrosis factor alpha (TNF-alpha), which migrates across the BBB and leads to inflammation and oxidative stress in brain, most likely contributing to the observed decline in cognition. In the current study, we measured levels of the antioxidant glutathione (GSH) in brains of mice injected intraparitoneally (i.p.) with ADR, as well as the levels and activities of several enzymes involved in brain GSH metabolism. We observed significantly decreased GSH levels, as well as altered GSH/GSSG ratio in brains of ADR treated mice relative to saline-treated controls. Also observed in brains of ADR treated mice were increased levels of glutathione peroxidase (GPx), glutathione-S-transferase (GST), and glutathione reductase (GR). We also observed increased activity of GPx, but a significant reduction in GST and GR activity in mice brain, 72 h post i.p. injection of ADR (20 mg/kg body weight). Furthermore, we used redox proteomics to identify specific proteins that are oxidized and/or have differential levels in mice brains as a result of a single i.p. injection of ADR. Visinin like protein 1 (VLP1), peptidyl prolyl isomerase 1 (Pin1), and syntaxin 1 (SYNT1) showed differential levels in ADR treated mice relative to saline-treated controls. Triose phosphate isomerase (TPI), enolase, and peroxiredoxin 1 (PRX-1) showed significantly increased specific carbonylation in ADR treated mice brain. These results further support the notion ADR induces oxidative stress in brain despite not crossing the BBB, and that antioxidant intervention may prevent ADR-induced cognitive dysfunction.
Collapse
Affiliation(s)
- G Joshi
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Díez-Vives C, Gay M, García-Matas S, Comellas F, Carrascal M, Abian J, Ortega-Aznar A, Cristòfol R, Sanfeliu C. Proteomic study of neuron and astrocyte cultures from senescence-accelerated mouse SAMP8 reveals degenerative changes. J Neurochem 2009; 111:945-55. [PMID: 19735447 DOI: 10.1111/j.1471-4159.2009.06374.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Senescence-accelerated prone (SAMP) strain 8 mice suffer an earlier development of cognitive age-related pathologies and a shorter life span than conventional mice. Protein alterations in astrocytes, in addition to those in neurons, may contribute to neurodegenerative damage. We applied proteomics techniques to study cell-specific early markers of brain aging-related degeneration in SAMP8. The two-dimensional protein expression patterns of the SAMP8 neuron and astrocyte cultures were compared with those obtained from senescence-accelerated resistant mouse strain 1 cultures. Differentially expressed spots were identified by matrix-assisted laser desorption/ionization-time of flight peptide map fingerprinting and database search. Proteins belonged to cell pathways of energy metabolism, biosynthesis, cell transduction and signaling, stress response, and the maintenance of cytoskeletal functions. Most of the changes were cell type specific. However, there was a general increase in cell transduction, signaling, and stress-related proteins and a decrease in cytoskeletal proteins. In addition, neurons showed an increased expression of proteins involved in biosynthetic pathways. A number of the protein alterations have been previously reported in the brain tissue proteome of SAMP8, aged brain or Alzheimer's disease brain. Alterations in neuron and astrocyte proteoma indicated that both cell types are involved in the brain degenerative changes of SAMP8 mice. However, network analysis suggests that neuronal changes are more complex and have a greater influence.
Collapse
Affiliation(s)
- Cristina Díez-Vives
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona, CSIC-IDIBAPS, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Neuroproteomics: understanding the molecular organization and complexity of the brain. Nat Rev Neurosci 2009; 10:635-46. [DOI: 10.1038/nrn2701] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Abstract
Olfactory bulbs (OBs) are one of the few brain areas, which show active neurogenesis and neuronal migration processes in adult rats. We constructed a proteome map of the 21 days old rat OBs and identified total 196 proteins, out of which 76 proteins were not reported earlier from rat brain. This includes 24 neuronal activity-specific proteins present at high levels, 7 of which are reported for the first time from OBs.
Collapse
Affiliation(s)
- Devendra Kumar Maurya
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research, Hyderabad, India
| | | | | |
Collapse
|
21
|
Badonnel K, Durieux D, Monnerie R, Grébert D, Salesse R, Caillol M, Baly C. Leptin-sensitive OBP-expressing mucous cells in rat olfactory epithelium: a novel target for olfaction-nutrition crosstalk? Cell Tissue Res 2009; 338:53-66. [PMID: 19688223 DOI: 10.1007/s00441-009-0846-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Accepted: 07/10/2009] [Indexed: 01/22/2023]
Abstract
Although odorant-binding proteins (OBP) are one of the most abundant classes of proteins in the mammalian olfactory mucus, they have only recently been ascribed a functional role in the detection of odorants by olfactory neurons. Among the three OBPs described in the rat, OBP-1f is mainly secreted by the lateral nasal glands (LNG) and Bowman's glands, and its expression is transcriptionally regulated by food deprivation in the olfactory mucosa, but not in LNG. Therefore, mucus composition might be locally regulated by hormones or molecules relevant to nutritional status. Our aim has been to investigate the mechanisms of such physiological regulation at the cellular level, through both the examination of OBP-1f synthesis sites in the olfactory mucosa and their putative regulation by leptin, a locally acting satiety hormone. Immunohistochemical observations have allowed the identification of a novel population of OBP-1f-secreting cells displaying morphological and functional characteristics similar to those of epithelial mucous cells. Ultrastructural analyses by both transmission and scanning electron microscopy has enabled a more complete cytoarchitectural characterization of these specialized olfactory mucous cells in their tissue environment. These globular cells are localized in discrete zones of the olfactory epithelium, mainly in the fourth turbinate, and are often scattered from the basal to the apical surface of the epithelium. They contain numerous small droplets of mucosubstances. Using an in-vitro-derived model of olfactory mucosa primary culture, we have been able to demonstrate that leptin increases the production of mucus by these cells, so that they constitute potential targets for the physiological modulation of mucus composition by nutritional cues.
Collapse
Affiliation(s)
- Karine Badonnel
- INRA, UMR1197 Neurobiologie de l'Olfaction et de la Prise Alimentaire, Jouy en Josas, France
| | | | | | | | | | | | | |
Collapse
|
22
|
Chakravarti B, Seshi B, Ratanaprayul W, Dalal N, Lin L, Raval A, Chakravarti DN. Proteome profiling of aging in mouse models: differential expression of proteins involved in metabolism, transport, and stress response in kidney. Proteomics 2009; 9:580-97. [PMID: 19184973 DOI: 10.1002/pmic.200700208] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Aging is a time-dependent complex biological phenomenon observed in various organs and organelles of all living organisms. To understand the molecular mechanism of age-associated functional loss in aging kidneys, we have analyzed the expression of proteins in the kidneys of young (19-22 wk) and old (24 months) C57/BL6 male mice using 2-DE followed by LC-MS/MS. We found that expression levels of 49 proteins were upregulated (p < or = 0.05), while that of only ten proteins were downregulated (p < or = 0.05) due to aging. The proteins identified belong to three broad functional categories: (i) metabolism (e.g., aldehyde dehydrogenase family, ATP synthase beta-subunit, malate dehydrogenase, NADH dehydrogenase (ubiquinone), hydroxy acid oxidase 2), (ii) transport (e.g., transferrin), and (iii) chaperone/stress response (e.g., Ig-binding protein, low density lipoprotein receptor-related protein associated protein 1, selenium-binding proteins (SBPs)). Some proteins with unknown functions were also identified as being differentially expressed. ATP synthase beta subunit, transferrin, fumarate hydratase, SBPs, and albumin are present in multiple forms, possibly arising due to proteolysis or PTMs. The above functional categories suggest specific mechanisms and pathways for age-related kidney degeneration.
Collapse
Affiliation(s)
- Bulbul Chakravarti
- Keck Graduate Institute of Applied Life Sciences, Claremont, CA 91711, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Owen JB, Di Domenico F, Sultana R, Perluigi M, Cini C, Pierce WM, Butterfield DA. Proteomics-determined differences in the concanavalin-A-fractionated proteome of hippocampus and inferior parietal lobule in subjects with Alzheimer's disease and mild cognitive impairment: implications for progression of AD. J Proteome Res 2009; 8:471-82. [PMID: 19072283 DOI: 10.1021/pr800667a] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia, comprising 60-80% of all reported cases, and currently affects 5.2 million Americans. AD is characterized pathologically by the accumulation of senile plaques (SPs), neurofibrillary tangles (NFTs), and synapse loss. The early stages of memory loss associated with AD have been studied in a condition known as amnestic mild cognitive impairment (MCI), arguably the earliest form of AD. In spite of extensive research across a variety of disciplines, the cause of AD remains elusive. Proteomics techniques have helped to advance knowledge about AD by identifying irregularities in protein expression and post-translational modifications (PTMs) in AD brain. Glycosylation is a less studied PTM with regards to AD and MCI. This PTM is important to study because glycosylation is involved in proper protein folding, protein anchoring to cell membranes, and the delivery of proteins to organelles, and these processes are impaired in AD. Concanavalin-A (Con-A) binds to N-linked glycoproteins, but hydrophobic sites on nonglycoproteins are also known to bind Con-A. To our knowledge, the present study is the first to examine Con-A-associated brain proteins in MCI and AD with focus on the hippocampus and inferior parietal lobule (IPL) brain regions. Proteins found in AD hippocampus with altered levels are glutamate dehydrogenase (GDH), glial fibrillary acidic protein (GFAP), tropomyosin 3 (TPM3), Rab GDP-dissociation inhibitor XAP-4 (XAP4), and heat shock protein 90 (HSP90). Proteins found with altered levels in AD IPL are alpha-enolase, gamma-enolase, and XAP-4. MCI hippocampal proteins with altered levels are dihydropyrimidase-2 (DRP2), glucose-regulated protein 78 (GRP-78), protein phosphatase related protein Sds-22 (Sds22), and GFAP and the only protein found with altered levels in MCI IPL was beta-synuclein. These results are discussed with reference to biochemical and pathological alterations in and progression of AD.
Collapse
Affiliation(s)
- Joshua B Owen
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506-0055, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Reed TT, Owen J, Pierce WM, Sebastian A, Sullivan PG, Butterfield DA. Proteomic identification of nitrated brain proteins in traumatic brain-injured rats treated postinjury with gamma-glutamylcysteine ethyl ester: Insights into the role of elevation of glutathione as a potential therapeutic strategy for traumatic brain injury. J Neurosci Res 2009; 87:408-17. [DOI: 10.1002/jnr.21872] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
Acupuncture inhibits ferric iron deposition and ferritin-heavy chain reduction in an MPTP-induced parkinsonism model. Neurosci Lett 2009; 450:92-6. [DOI: 10.1016/j.neulet.2008.11.049] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 10/23/2008] [Accepted: 11/16/2008] [Indexed: 11/20/2022]
|
26
|
Tilton F, Tilton SC, Bammler TK, Beyer R, Farin F, Stapleton PL, Gallagher EP. Transcriptional biomarkers and mechanisms of copper-induced olfactory injury in zebrafish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2008; 42:9404-11. [PMID: 19174923 PMCID: PMC3321378 DOI: 10.1021/es801636v] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Metals such as copper disrupt olfactory function in fish. Unfortunately, little is understood of the molecular consequences of copper olfactory impairment, thus hindering the development of relevant diagnostic tools of olfactory injury. To address this critical data gap, we analyzed gene expression within olfactory tissues of adult zebrafish exposed to CuCl2 (6, 16, 40 ppb) for 24 h. Transcriptional markers of copper impairment within the entire olfactory system were identified and specific genes of interest (e.g., S100a, parvalbumin 8, olfactory marker protein, and calbindin 2-like protein) were confirmed with quantitative real-time PCR. In addition, we performed gene set analysis (GSA) using both a priori and custom pathways of gene sets specifically targeting the olfactory signal transduction (OST) pathway. These analyses revealed down-regulated gene sets related to calcium channels and ion transport, g-proteins, and olfactory receptors. Collectively, these data demonstrate that copper causes a depression of transcription of key genes within the OST pathway and elsewhere within olfactory tissues, likely resulting in an olfactory system less responsive to odorants. Further, these data provide a mechanistic explanation in support of earlier studies of functional olfactory impairment in fish following copper exposure.
Collapse
Affiliation(s)
- Fred Tilton
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Sultana R, Butterfield DA. Redox Proteomics Analysis of Oxidative Modified Brain Proteins in Alzheimer's Disease and Mild Cognitive Impairment: Insights into the Progression of This Dementing Disorder. Clin Proteomics 2008. [DOI: 10.1002/9783527622153.ch23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
28
|
Vaishnav RA, Getchell ML, Huang L, Hersh MA, Stromberg AJ, Getchell TV. Cellular and molecular characterization of oxidative stress in olfactory epithelium of Harlequin mutant mouse. J Neurosci Res 2008; 86:165-82. [PMID: 17868149 DOI: 10.1002/jnr.21464] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Oxidative stress in the olfactory system is a major factor associated with age-related olfactory impairment, although the mechanisms by which this occurs are not completely understood. The Harlequin mutant mouse (Hq/Y), which carries an X-linked recessive mutation in the Aifm1 gene, is a model of progressive oxidative stress-induced neurodegeneration in the cerebellum and retina. To determine whether the Hq/Y mutant mouse is a suitable model of oxidative stress-associated olfactory aging, we investigated cellular and molecular changes in the olfactory epithelium (OE) and olfactory bulb (OB) of 6-month-old male Hq/Y mice compared to those in sex-matched littermate controls (+/Y) and in age- and sex-matched C57BL/6 mice. Immunoreactivity for apoptosis-inducing factor, the protein product of Aifm1, was localized in mature olfactory sensory neurons (mOSNs) in +/Y mice but was rarely detected in Hq/Y mice. Hq/Y mice also exhibited increased lipofuscin autofluorescence and increased immunoreactivity for an oxidative DNA/RNA damage marker in mOSNs and in mitral/tufted cells in the OB and an increased number of cleaved caspase-3 immunoreactive apoptotic cells in the OE. Microarray analysis demonstrated that Aifm1 expression was down-regulated by 80% in the OE of Hq/Y mice compared to that in +/Y mice. Most significantly, regulated genes were classified into functional categories of cell signaling/apoptosis/cell cycle, oxidative stress/aging, and cytoskeleton/extracellular matrix/transport-associated. Analysis with EASE software indicated that the functional categories significantly overrepresented in Hq/Y mice included up-regulated mitochondrial genes and down-regulated cytoskeletal organization- and neurogenesis-related genes. Our results strongly support the Hq/Y mutant mouse being a novel model for mechanistic studies of oxidative stress-associated olfactory aging.
Collapse
Affiliation(s)
- Radhika A Vaishnav
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | | | | | | | | | | |
Collapse
|
29
|
Barbour J, Neuhaus EM, Piechura H, Stoepel N, Mashukova A, Brunert D, Sitek B, Stühler K, Meyer HE, Hatt H, Warscheid B. New insight into stimulus-induced plasticity of the olfactory epithelium in Mus musculus by quantitative proteomics. J Proteome Res 2008; 7:1594-605. [PMID: 18336002 DOI: 10.1021/pr7005796] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The olfactory system is exposed to a plethora of chemical compounds throughout an organism's lifespan. Anticipation of stimuli and construction of appropriate neural filters present a significant challenge. This may be addressed via modulation of the protein composition of the sensory epithelium in response to environmental conditions. To reveal the mechanisms governing these changes, we employed a comprehensive quantitative proteomics strategy. Two groups of juvenile mice were treated with either pulsed or continuous application of octanal. After 20 days of treatment, we performed a behavioral study and conducted electrophysiological recordings from the olfactory epithelium (OE). Both treated groups demonstrated peripheral desensitization to octanal; however, only the 'continuous' group exhibited habituation. To obtain novel insight into the molecular mechanisms underpinning the peripheral desensitization to octanal, the OE proteomes of octanal-treated mice versus control were quantitatively analyzed using two-dimensional difference gel electrophoresis. We identified several significantly regulated proteins that were functionally classified as calcium-binding proteins, cytoskeletal proteins, and lipocalins. The calcium-binding proteins and cytoskeletal proteins were up-regulated in the 'pulsed' group, whereas in the 'continuous' group, four lipocalins were significantly down-regulated. Uniquely, the lipocalin odorant-binding protein Ia was drastically down-regulated in both groups. The identified proteins reflect changes throughout the entire OE, corresponding to changes in neuronal, non-neuronal, and pericellular processes. We report the regulation of several promising candidates for the investigation of odorant-induced changes of the OE. Among these proteins are different lipocalins, which seem to play a crucial role in the regulation of the sensitivity of the olfactory system.
Collapse
Affiliation(s)
- Jon Barbour
- Medizinisches Proteom-Center, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Detection of carbonylated proteins in two-dimensional sodium dodecyl sulfate polyacrylamide gel electrophoresis separations. Methods Mol Biol 2008; 476:153-63. [PMID: 19157015 DOI: 10.1007/978-1-59745-129-1_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Protein carbonyls are an index of protein oxidation which, in turn, reflects the interplay of oxidative stress and degradation of oxidatively modified proteins. Protein carbonyls are increased in brain proteins in aging and age-related neurodegenerative disorders, including Alzheimer's disease. In this chapter, we outline methods to detect protein carbonyls following two dimensional-based separation of brain proteins.
Collapse
|
31
|
Weinreb O, Amit T, Youdim MBH. A novel approach of proteomics and transcriptomics to study the mechanism of action of the antioxidant-iron chelator green tea polyphenol (-)-epigallocatechin-3-gallate. Free Radic Biol Med 2007; 43:546-56. [PMID: 17640565 DOI: 10.1016/j.freeradbiomed.2007.05.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 04/25/2007] [Accepted: 05/07/2007] [Indexed: 10/23/2022]
Abstract
Previous findings suggest that the antioxidant-iron chelator green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG) may have a neurorescue impact in aging and neurodegenerative diseases to retard or even reverse the accelerated rate of neuronal degeneration. The present study sought a deeper elucidation of the molecular neurorescue activity of EGCG in a progressive neurotoxic model of long-term serum deprivation of human SH-SY5Y neuroblastoma cells. In this model, proteomic analysis revealed that EGCG (0.1-1 microM) affected the expression levels of diverse proteins, including proteins related to cytoskeletal components, metabolism, heat shock, and binding. EGCG induced the levels of cytoskeletal proteins, such as beta tubulin IV and tropomyosin 3, playing a role in facilitating cell assembly. In accordance, EGCG increased the levels of the binding protein 14-3-3 gamma, involved in cytoskeletal regulation and signal transduction pathways in neurons. Additionally, EGCG decreased protein levels and mRNA expression of the beta subunit of the enzyme prolyl 4-hydroxylase, which belongs to a family of iron-oxygen sensors of hypoxia-inducible factor (HIF) prolyl hydroxylases that negatively regulate the stability and degradation of several proteins involved in cell survival and differentiation. Accordingly, EGCG decreased protein levels of two molecular chaperones that were associated with HIF regulation, the immunoglobulin-heavy-chain binding protein and the heat shock protein 90 beta. Thus, the present study sheds some light on the antioxidative-iron chelating activities of EGCG underlying its neuroprotective/neurorescue mechanism of action, further suggesting a potential neurodegenerative-modifying effect for EGCG.
Collapse
Affiliation(s)
- Orly Weinreb
- Eve Topf and USA National Parkinson Foundation Centers of Excellence for Neurodegenerative Diseases Research and Department of Pharmacology, Rappaport Family Research Institute, Technion-Faculty of Medicine, Haifa 31096, Israel
| | | | | |
Collapse
|
32
|
Groebe K, Krause F, Kunstmann B, Unterluggauer H, Reifschneider NH, Scheckhuber CQ, Sastri C, Stegmann W, Wozny W, Schwall GP, Poznanović S, Dencher NA, Jansen-Dürr P, Osiewacz HD, Schrattenholz A. Differential proteomic profiling of mitochondria from Podospora anserina, rat and human reveals distinct patterns of age-related oxidative changes. Exp Gerontol 2007; 42:887-98. [PMID: 17689904 DOI: 10.1016/j.exger.2007.07.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Revised: 06/18/2007] [Accepted: 07/06/2007] [Indexed: 01/07/2023]
Abstract
According to the 'free radical theory of ageing', the generation and accumulation of reactive oxygen species are key events during ageing of biological systems. Mitochondria are a major source of ROS and prominent targets for ROS-induced damage. Whereas mitochondrial DNA and membranes were shown to be oxidatively modified with ageing, mitochondrial protein oxidation is not well understood. The purpose of this study was an unbiased investigation of age-related changes in mitochondrial proteins and the molecular pathways by which ROS-induced protein oxidation may disturb cellular homeostasis. In a differential comparison of mitochondrial proteins from young and senescent strains of the fungal ageing model Podospora anserina, from brains of young (5 months) vs. older rats (17 and 31 months), and human cells, with normal and chemically accelerated in vitro ageing, we found certain redundant posttranslationally modified isoforms of subunits of ATP synthase affected across all three species. These appear to represent general susceptible hot spot targets for oxidative chemical changes of proteins accumulating during ageing, and potentially initiating various age-related pathologies and processes. This type of modification is discussed using the example of SAM-dependent O-methyltransferase from P. anserina (PaMTH1), which surprisingly was found to be enriched in mitochondrial preparations of senescent cultures.
Collapse
|
33
|
Mu J, Xie P, Yang ZS, Yang DL, Lv FJ, Luo TY, Li Y. Neurogenesis and major depression: implications from proteomic analyses of hippocampal proteins in a rat depression model. Neurosci Lett 2007; 416:252-6. [PMID: 17368938 DOI: 10.1016/j.neulet.2007.01.067] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 12/28/2006] [Accepted: 01/20/2007] [Indexed: 11/24/2022]
Abstract
Major depression is one of the most disabling disorders. Yet, the pathogenesis of this mental disorder is poorly understood. Hippocampus is generally believed to be associated with pathogenesis of depression. In this study, we adopted a proteomic approach to examine possible alterations of protein expression in the hippocampus of a rat depression model. Our results suggest that neurogenesis in hippocampus may play an important role in the pathogenesis of major depression.
Collapse
Affiliation(s)
- Jun Mu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, The Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing 400016, China
| | | | | | | | | | | | | |
Collapse
|
34
|
Vaishnav RA, Getchell ML, Poon HF, Barnett KR, Hunter SA, Pierce WM, Klein JB, Butterfield DA, Getchell TV. Oxidative stress in the aging murine olfactory bulb: redox proteomics and cellular localization. J Neurosci Res 2007; 85:373-85. [PMID: 17131389 DOI: 10.1002/jnr.21130] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A recent proteomics analysis from our laboratory demonstrated that several oxidative stress response proteins showed significant changes in steady-state levels in olfactory bulbs (OBs) of 20- vs. 1.5-month-old mice. Oxidative stress may result in protein oxidation. In this study, we investigated two forms of protein oxidative modification in murine OBs: carbonylation and nitration. Redox proteomics with two-dimensional gel electrophoresis, Western blotting, protein digestion, and mass spectrometry was used to quantify total and specific protein carbonylation and to identify differentially carbonylated proteins and determine the carbonylation status of previously identified proteins in OBs of 1.5- and 20-month-old mice. Immunohistochemistry was used to demonstrate the relative intensity and localization of protein nitration in OBs of 1.5-, 6-, and 20-month-old mice. Total protein carbonylation was significantly greater in OBs of 20- vs. 1.5-month-old mice. Aldolase 1 (ALDO1) showed significantly more carbonylation in OBs from 20- vs. 1.5-month-old mice; heat shock protein 9A and dihydropyrimidinase-like 2 showed significantly less. Several previously investigated proteins were also carbonylated, including ferritin heavy chain (FTH). Nitration, identified by 3-nitrotyrosine immunoreactivity, was least abundant at 1.5 months, intermediate at 6 months, and greatest at 20 months and was localized primarily in blood vessels. Proteins that were specific targets of oxidation were also localized: ALDO1 in astrocytes of the granule cell layer and FTH in mitral/tufted cells. These results indicate that specific carbonylated proteins, including those in astrocytes and mitral/tufted neurons, and nitrated proteins in the vasculature are molecular substrates of age-related olfactory dysfunction.
Collapse
Affiliation(s)
- Radhika A Vaishnav
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Hasegawa H, Takano H, Kohro T, Ueda K, Niitsuma Y, Aburatani H, Komuro I. Amelioration of hypertensive heart failure by amlodipine may occur via antioxidative effects. Hypertens Res 2007; 29:719-29. [PMID: 17249528 DOI: 10.1291/hypres.29.719] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Although recent clinical studies have suggested that long-acting calcium channel blockers (CCBs) have beneficial effects on heart failure, the precise mechanism is unknown. In this study, Dahl salt-sensitive rats fed a high salt diet were treated with the long-acting CCB amlodipine, the low-molecular-weight membrane permeable superoxide dismutase mimetic 4-hydroxy-2,2,6,6-tetramethyl piperidinoxyl (Tempol), or saline from 11 weeks after birth. The cardiac geometry and function, and gene expression profiles were determined at 17 weeks. Dahl salt-sensitive rats fed a high salt diet followed by saline as a non-treatment control (HS group) showed a marked increase in blood pressure and developed concentric hypertrophy at 11 weeks, followed by left ventricular (LV) dilation and congestive heart failure by 17 weeks. The treatment with amlodipine (AMLO group) or Tempol (TEMP group) significantly inhibited the development of LV hypertrophy and cardiac dysfunction. Analysis using an Affymetrix GeneChip U34 revealed that the expression levels of 195 genes were changed by the treatment with amlodipine. Among these 195 genes, 110 genes were increased in HS rats and decreased in AMLO rats. And of these 110 genes, 54 genes were also decreased in TEMP rats. In contrast, 85 genes were decreased in HS rats and increased in AMLO rats. Of these 85 genes, 38 genes were also increased in TEMP rats. Approximately 48% of the genes were changed in similar fashion in AMLO and TEMP rats, suggesting that amlodipine shows beneficial effects on heart failure mainly via antioxidative mechanisms.
Collapse
Affiliation(s)
- Hiroshi Hasegawa
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Weinreb O, Drigues N, Sagi Y, Reznick AZ, Amit T, Youdim MBH. The application of proteomics and genomics to the study of age-related neurodegeneration and neuroprotection. Antioxid Redox Signal 2007; 9:169-79. [PMID: 17115941 DOI: 10.1089/ars.2007.9.169] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The present study aimed to acquire more information on aging-related alterations, using proteomic and genomic analyses of hippocampus from young (8 months) and old (27 months) rats. In the old rats, the proteomic analysis identified changes in proteins related to the iron-mediated oxidative stress (OS) pathway, including reduction in antioxidant enzymes (e.g., peroxiredoxin, cytochrome c oxidase) and induction of ferritin. Furthermore, the neurofilament light peptide, associated with neurodegenerative processes, was enhanced and binding/ chaperone proteins were altered in old vs. young rats. At the genes levels, significant molecular changes related to neurodegeneration were identified in aged rat hippocampus. Thus, the effects of the potent neuroprotective compounds, the anti-Parkinson drug, rasagiline and the anti-Alzheimer drug, ladostigil (1 mg/kg, for 30 days) on gene expression in the hippocampus were further investigated. Both drugs reversed the effect of aging on the expression of various mitochondrial and key regulator genes involved in neurodegeneration, cell survival, synaptogenesis, oxidation, and metabolism. These results support the hypothesis that OS and mitochondrial dysfunction may play a pivotal role in aging and age-associated neurodegenerative diseases, and can serve as potential clinical targets for future therapy.
Collapse
Affiliation(s)
- Orly Weinreb
- Eve Topf Center of Excellence for Neurodegenerative Diseases Research and Department of Pharmacology, Rappaport Family Research Institute, Faculty of Medicine, Technion, Haifa, Israel
| | | | | | | | | | | |
Collapse
|
37
|
Opii WO, Joshi G, Head E, William Milgram N, Muggenburg BA, Klein JB, Pierce WM, Cotman CW, Allan Butterfield D. Proteomic identification of brain proteins in the canine model of human aging following a long-term treatment with antioxidants and a program of behavioral enrichment: relevance to Alzheimer's disease. Neurobiol Aging 2006; 29:51-70. [PMID: 17055614 PMCID: PMC2203613 DOI: 10.1016/j.neurobiolaging.2006.09.012] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 09/06/2006] [Accepted: 09/14/2006] [Indexed: 01/28/2023]
Abstract
Aging and age-related disorders such as Alzheimer's disease (AD) are usually accompanied by oxidative stress as one of the main mechanisms contributing to neurodegeneration and cognitive decline. Aging canines develop cognitive dysfunction and neuropathology similar to those seen in humans, and the use of antioxidants results in reductions in oxidative damage and in improvement in cognitive function in this canine model of human aging. In the present study, the effect of a long-term treatment with an antioxidant-fortified diet and a program of behavioral enrichment on oxidative damage was studied in aged canines. To identify the neurobiological mechanisms underlying these treatment effects, the parietal cortex from 23 beagle dogs (8.1-12.4 years) were treated for 2.8 years in one of four treatment groups: i.e., control food-control behavioral enrichment (CC); control food-behavioral enrichment (CE); antioxidant food-control behavioral enrichment (CA); enriched environment-antioxidant-fortified food (EA). We analyzed the levels of the oxidative stress biomarkers, i.e., protein carbonyls, 3-nitrotyrosine (3-NT), and the lipid peroxidation product, 4-hydroxynonenal (HNE), and observed a decrease in their levels on all treatments when compared to control, with the most significant effects found in the combined treatment, EA. Since EA treatment was most effective, we also carried out a comparative proteomics study to identify specific brain proteins that were differentially expressed and used a parallel redox proteomics approach to identify specific brain proteins that were less oxidized following EA. The specific protein carbonyl levels of glutamate dehydrogenase [NAD (P)], glyceraldehyde-3-phosphate dehydrogenase (GAPDH), alpha-enolase, neurofilament triplet L protein, glutathione-S-transferase (GST) and fascin actin bundling protein were significantly reduced in brain of EA-treated dogs compared to control. We also observed significant increases in expression of Cu/Zn superoxide dismutase, fructose-bisphosphate aldolase C, creatine kinase, glutamate dehydrogenase and glyceraldehyde-3-phosphate dehydrogenase. The increased expression of these proteins and in particular Cu/Zn SOD correlated with improved cognitive function. In addition, there was a significant increase in the enzymatic activities of glutathione-S-transferase (GST) and total superoxide dismutase (SOD), and significant increase in the protein levels of heme oxygenase (HO-1) in EA treated dogs compared to control. These findings suggest that the combined treatment reduces the levels of oxidative damage and improves the antioxidant reserve systems in the aging canine brain, and may contribute to improvements in learning and memory. These observations provide insights into a possible neurobiological mechanism underlying the effects of the combined treatment. These results support the combination treatments as a possible therapeutic approach that could be translated to the aging human population who are at risk for age-related neurodegenerative disorders, including Alzheimer's disease.
Collapse
Affiliation(s)
- Wycliffe. O. Opii
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington KY 40506-0055
| | - Gururaj Joshi
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington KY 40506-0055
| | - Elizabeth Head
- Institute for Brain Aging and Dementia, Department of Neurology, University of California, Irvine, California, 92697-4540
| | - N William Milgram
- Division of Life Sciences, University of Toronto, Toronto, Canada, M1C 1A4
| | | | - Jon B. Klein
- Department of Medicine, Kidney Disease Program, University of Louisville, Louisville, KY
| | | | - Carl. W. Cotman
- Institute for Brain Aging and Dementia, Department of Neurology, University of California, Irvine, California, 92697-4540
| | - D Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington KY 40506-0055
- *Address Correspondence to: Prof. D. Allan Butterfield, Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA, Ph: 859-257-3184; FAX: 859-257-5876; E-Mail:
| |
Collapse
|
38
|
Stehle JR, Weeks ME, Lin K, Willingham MC, Hicks AM, Timms JF, Cui Z. Mass spectrometry identification of circulating alpha-1-B glycoprotein, increased in aged female C57BL/6 mice. Biochim Biophys Acta Gen Subj 2006; 1770:79-86. [PMID: 16945486 DOI: 10.1016/j.bbagen.2006.06.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Revised: 06/12/2006] [Accepted: 06/13/2006] [Indexed: 11/27/2022]
Abstract
In this study, we surveyed the profiles of mouse circulating proteins by 2-dimensional SDS-PAGE in different strains, sexes and ages. Among visible protein spots on 2-D gels with silver-staining, we identified a unique set of 7 seemingly-related proteins whose levels were consistently elevated in older C57BL/6 female mice. This set of 7 proteins was absent in C57BL/6 males or in BALB/c mice of either sex of any age. When C57BL/6 female mice were crossed with BALB/c males, the age-related increase of these proteins became sporadic and not linear in the F1 offspring. All 7 spots of this protein group were picked and subjected to identification by mass spectrometric analysis after tryptic digestion. The results showed that all 7 spots were different isoforms of alpha(1)B-glycoprotein with different degrees of post-translational modifications, such as phosphorylation. These results suggest that alpha(1)B-glycoprotein changes in mice in a sex and age dependent manner.
Collapse
Affiliation(s)
- John R Stehle
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Rawson NE, LaMantia AS. A speculative essay on retinoic acid regulation of neural stem cells in the developing and aging olfactory system. Exp Gerontol 2006; 42:46-53. [PMID: 16860961 DOI: 10.1016/j.exger.2006.05.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Accepted: 05/19/2006] [Indexed: 11/22/2022]
Abstract
Circulating signals like the acidic derivative of vitamin A: retinoic acid (RA) may regulate resident stem cells in the adult nervous system, particularly in the olfactory pathway. RA is an essential factor for inducing neural stem or precursor cells that give rise to olfactory receptor neurons (ORNs) and olfactory bulb (OB) interneurons (OBINs) during embryonic development. Similar precursors in the adult brain constantly generate new ORNs and OBINs, and embryonic signaling pathways, like that via RA, may be retained or reactivated for this purpose. We have shown that RA regulates neural precursors in the embryonic and adult olfactory pathway. Moreover, RA administration after olfactory system damage stimulates an immune response and yields a more rapid recovery of olfactory-guided behavior. We suggest that olfactory integrity may be maintained by RA-mediated regulation of neurogenesis as well as local immune responses, and that aging compromises these mechanisms. The chemical senses, particularly olfaction, decline in aged individuals, and RA (via vitamin A) levels may also decline, perhaps due to changes in appetite and food intake. This synergy may result in a high prevalence of olfactory pathology in aged individuals.
Collapse
Affiliation(s)
- N E Rawson
- Monell Chemical Senses Center, Philadelphia, PA, USA
| | | |
Collapse
|
40
|
Poon HF, Vaishnav RA, Getchell TV, Getchell ML, Butterfield DA. Quantitative proteomics analysis of differential protein expression and oxidative modification of specific proteins in the brains of old mice. Neurobiol Aging 2006; 27:1010-9. [PMID: 15979213 DOI: 10.1016/j.neurobiolaging.2005.05.006] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2004] [Revised: 03/26/2005] [Accepted: 05/07/2005] [Indexed: 11/26/2022]
Abstract
The brain is susceptible to oxidative stress, which is associated with age-related brain dysfunction, because of its high content of peroxidizable unsaturated fatty acids, high oxygen consumption per unit weight, high content of key components for oxidative damage, and the relative scarcity of antioxidant defense systems. Protein oxidation, which results in functional disruption, is not random but appears to be associated with increased oxidation in specific proteins. By using a proteomics approach, we have compared the protein levels and specific protein carbonyl levels, an index of oxidative damage in the brains of old mice, to these parameters in the brains of young mice and have identified specific proteins that are altered as a function of aging. We show here that the expression levels of dihydropyrimidinase-like 2 (DRP2), alpha-enolase (ENO1), dynamin-1 (DNM1), and lactate dehydrogenase 2 (LDH2) were significantly increased in the brains of old versus young mice; the expression levels of three unidentified proteins were significantly decreased. The specific carbonyl levels of beta-actin (ACTB), glutamine synthase (GS), and neurofilament 66 (NF-66) as well as a novel protein were significantly increased, indicating protein oxidation, in the brains of old versus young mice. These results were validated by immunochemistry. In addition, enzyme activity assays demonstrated that oxidation was associated with decreased GS activity, while the activity of lactate dehydrogenase was unchanged in spite of an up-regulation of LDH2 levels. Several of the up-regulated and oxidized proteins in the brains of old mice identified in this report are known to be oxidized in neurodegenerative diseases as well, suggesting that these proteins may be particularly susceptible to processes associated with neurodegeneration. Our results establish an initial basis for understanding protein alterations that may lead to age-related cellular dysfunction in the brain.
Collapse
Affiliation(s)
- H Fai Poon
- Department of Chemistry, Center of Mambrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
Olfactory loss is a common age-related complaint that may be caused by changes in the anatomy of the structures required for olfaction (for example, loss of olfactory receptor cells) or in the environment surrounding the receptor cell (for example, altered nasal mucus composition). However, aging, as well as age-related diseases and medications, may also alter the distribution, density, or function of specific receptor proteins, ion channels, or signaling molecules that affect the ability of neural elements throughout the olfactory pathway to signal and process odorant information. Although a great deal has been learned about the prevalence and nature of age-related olfactory loss, we are just beginning to explore avenues to prevent or alleviate this sensory deficit. Some studies suggest that, rather than being a necessary outcome of aging, age-associated factors such as chronic diseases, medications, and dental and sinus problems are the primary culprits in causing olfactory impairment. This idea suggests optimism in that, as we address these other age-related health issues, the prevalence of olfactory loss will lessen as well.
Collapse
Affiliation(s)
- Nancy E Rawson
- Monell Chemical Senses Center, Philadelphia, PA 19104, USA.
| |
Collapse
|