1
|
Yang YC, Chien Y, Yarmishyn AA, Lim LY, Tsai HY, Kuo WC, Tsai PH, Yang SH, Hong SI, Chen SJ, Hwang DK, Yang YP, Chiou SH. Inhibition of oxidative stress-induced epithelial-mesenchymal transition in retinal pigment epithelial cells of age-related macular degeneration model by suppressing ERK activation. J Adv Res 2024; 60:141-157. [PMID: 37328058 PMCID: PMC11156608 DOI: 10.1016/j.jare.2023.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/05/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023] Open
Abstract
INTRODUCTION Epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is related to the pathogenesis of various retinopathies including age-related macular degeneration (AMD). Oxidative stress is the major factor that induces degeneration of RPE cells associated with the etiology of AMD. OBJECTIVES Sodium iodate (NaIO3) generates intracellular reactive oxygen species (ROS) and is widely used to establish a model of AMD due to the selective induction of retinal degeneration. This study was performed to clarify the effects of multiple NaIO3-stimulated signaling pathways on EMT in RPE cells. METHODS The EMT characteristics in NaIO3-treated human ARPE-19 cells and RPE cells of the mouse eyes were analyzed. Multiple oxidative stress-induced modulators were investigated and the effects of pre-treatment with Ca2+ chelator, extracellular signal-related kinase (ERK) inhibitor, or epidermal growth factor receptor (EGFR) inhibitor on NaIO3-induced EMT were determined. The efficacy of post-treatment with ERK inhibitor on the regulation of NaIO3-induced signaling pathways was dissected and its role in retinal thickness and morphology was evaluated by using histological cross-sections and spectral domain optical coherence tomography. RESULTS We found that NaIO3 induced EMT in ARPE-19 cells and in RPE cells of the mouse eyes. The intracellular ROS, Ca2+, endoplasmic reticulum (ER) stress marker, phospho-ERK, and phospho-EGFR were increased in NaIO3-stimulated cells. Our results showed that pre-treatment with Ca2+ chelator, ERK inhibitor, or EGFR inhibitor decreased NaIO3-induced EMT, interestingly, the inhibition of ERK displayed the most prominent effect. Furthermore, post-treatment with FR180204, a specific ERK inhibitor, reduced intracellular ROS and Ca2+ levels, downregulated phospho-EGFR and ER stress marker, attenuated EMT of RPE cells, and prevented structural disorder of the retina induced by NaIO3. CONCLUSIONS ERK is a crucial regulator of multiple NaIO3-induced signaling pathways that coordinate EMT program in RPE cells. Inhibition of ERK may be a potential therapeutic strategy for the treatment of AMD.
Collapse
Affiliation(s)
- Ya-Chi Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Aliaksandr A Yarmishyn
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Lee-Yieng Lim
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Hao-Yu Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Wen-Chuan Kuo
- Institute of Biophotonics, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Sheng-Hsien Yang
- Institute of Biophotonics, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Shao-I Hong
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Shih-Jen Chen
- College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - De-Kuang Hwang
- College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan.
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan; College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan; Genomic Research Center, Academia Sinica, Taipei 115024, Taiwan.
| |
Collapse
|
2
|
Pawłowska M, Mila-Kierzenkowska C, Szczegielniak J, Woźniak A. Oxidative Stress in Parasitic Diseases-Reactive Oxygen Species as Mediators of Interactions between the Host and the Parasites. Antioxidants (Basel) 2023; 13:38. [PMID: 38247462 PMCID: PMC10812656 DOI: 10.3390/antiox13010038] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Oxidative stress plays a significant role in the development and course of parasitic infections, both in the attacked host organism and the parasite organism struggling to survive. The host uses large amounts of reactive oxygen species (ROS), mainly superoxide anion (O2•-) and hydrogen peroxide (H2O2), to fight the developing parasitic disease. On the other hand, the parasite develops the most effective defense mechanisms and resistance to the effects of ROS and strives to survive in the host organism it has colonized, using the resources and living environment available for its development and causing the host's weakening. The paper reviews the literature on the role of oxidative stress in parasitic diseases, which are the most critical epidemiological problem worldwide. The most common parasitosis in the world is malaria, with 300-500 million new cases and about 1 million deaths reported annually. In Europe and Poland, the essential problem is intestinal parasites. Due to a parasitic infection, the concentration of antioxidants in the host decreases, and the concentration of products of cellular components oxidation increases. In response to the increased number of reactive oxygen species attacking it, the parasites have developed effective defense mechanisms, including primarily the action of antioxidant enzymes, especially superoxide dismutase and nicotinamide adenine dinucleotide phosphate hydrogen (NADPH)-dependent complexes glutathione and thioredoxin.
Collapse
Affiliation(s)
- Marta Pawłowska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland; (C.M.-K.); (A.W.)
| | - Celestyna Mila-Kierzenkowska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland; (C.M.-K.); (A.W.)
| | - Jan Szczegielniak
- Physiotherapy Department, Faculty of Physical Education and Physiotherapy, Opole University of Technology, 45-758 Opole, Poland;
- Ministry of Internal Affairs and Administration’s Specialist Hospital of St. John Paul II, 48-340 Glucholazy, Poland
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-092 Bydgoszcz, Poland; (C.M.-K.); (A.W.)
| |
Collapse
|
3
|
Burtscher J, Mallet RT, Pialoux V, Millet GP, Burtscher M. Adaptive Responses to Hypoxia and/or Hyperoxia in Humans. Antioxid Redox Signal 2022; 37:887-912. [PMID: 35102747 DOI: 10.1089/ars.2021.0280] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Significance: Oxygen is indispensable for aerobic life, but its utilization exposes cells and tissues to oxidative stress; thus, tight regulation of cellular, tissue, and systemic oxygen concentrations is crucial. Here, we review the current understanding of how the human organism (mal-)adapts to low (hypoxia) and high (hyperoxia) oxygen levels and how these adaptations may be harnessed as therapeutic or performance enhancing strategies at the systemic level. Recent Advances: Hyperbaric oxygen therapy is already a cornerstone of modern medicine, and the application of mild hypoxia, that is, hypoxia conditioning (HC), to strengthen the resilience of organs or the whole body to severe hypoxic insults is an important preparation for high-altitude sojourns or to protect the cardiovascular system from hypoxic/ischemic damage. Many other applications of adaptations to hypo- and/or hyperoxia are only just emerging. HC-sometimes in combination with hyperoxic interventions-is gaining traction for the treatment of chronic diseases, including numerous neurological disorders, and for performance enhancement. Critical Issues: The dose- and intensity-dependent effects of varying oxygen concentrations render hypoxia- and/or hyperoxia-based interventions potentially highly beneficial, yet hazardous, although the risks versus benefits are as yet ill-defined. Future Directions: The field of low and high oxygen conditioning is expanding rapidly, and novel applications are increasingly recognized, for example, the modulation of aging processes, mood disorders, or metabolic diseases. To advance hypoxia/hyperoxia conditioning to clinical applications, more research on the effects of the intensity, duration, and frequency of altered oxygen concentrations, as well as on individual vulnerabilities to such interventions, is paramount. Antioxid. Redox Signal. 37, 887-912.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.,Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Vincent Pialoux
- Inter-University Laboratory of Human Movement Biology EA7424, University Claude Bernard Lyon 1, University of Lyon, Lyon, France
| | - Grégoire P Millet
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.,Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Ghoweri AO, Gagolewicz P, Frazier HN, Gant JC, Andrew RD, Bennett BM, Thibault O. Neuronal Calcium Imaging, Excitability, and Plasticity Changes in the Aldh2-/- Mouse Model of Sporadic Alzheimer's Disease. J Alzheimers Dis 2021; 77:1623-1637. [PMID: 32925058 PMCID: PMC7683088 DOI: 10.3233/jad-200617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background: Dysregulated signaling in neurons and astrocytes participates in pathophysiological alterations seen in the Alzheimer’s disease brain, including increases in amyloid-β, hyperphosphorylated tau, inflammation, calcium dysregulation, and oxidative stress. These are often noted prior to the development of behavioral, cognitive, and non-cognitive deficits. However, the extent to which these pathological changes function together or independently is unclear. Objective: Little is known about the temporal relationship between calcium dysregulation and oxidative stress, as some reports suggest that dysregulated calcium promotes increased formation of reactive oxygen species, while others support the opposite. Prior work has quantified several key outcome measures associated with oxidative stress in aldehyde dehydrogenase 2 knockout (Aldh2–/–) mice, a non-transgenic model of sporadic Alzheimer’s disease. Methods: Here, we tested the hypothesis that early oxidative stress can promote calcium dysregulation across aging by measuring calcium-dependent processes using electrophysiological and imaging methods and focusing on the afterhyperpolarization (AHP), synaptic activation, somatic calcium, and long-term potentiation in the Aldh2–/– mouse. Results: Our results show a significant age-related decrease in the AHP along with an increase in the slow AHP amplitude in Aldh2–/– animals. Measures of synaptic excitability were unaltered, although significant reductions in long-term potentiation maintenance were noted in the Aldh2–/– animals compared to wild-type. Conclusion: With so few changes in calcium and calcium-dependent processes in an animal model that shows significant increases in HNE adducts, Aβ, p-tau, and activated caspases across age, the current findings do not support a direct link between neuronal calcium dysregulation and uncontrolled oxidative stress.
Collapse
Affiliation(s)
- Adam O Ghoweri
- Pharmacology and Nutritional Sciences University of Kentucky, University of Kentucky Medical Center, Lexington, KY, USA
| | - Peter Gagolewicz
- Biomedical and Molecular Sciences and Centre for Neuroscience Studies, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Hilaree N Frazier
- Pharmacology and Nutritional Sciences University of Kentucky, University of Kentucky Medical Center, Lexington, KY, USA
| | - John C Gant
- Pharmacology and Nutritional Sciences University of Kentucky, University of Kentucky Medical Center, Lexington, KY, USA
| | - R David Andrew
- Biomedical and Molecular Sciences and Centre for Neuroscience Studies, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Brian M Bennett
- Biomedical and Molecular Sciences and Centre for Neuroscience Studies, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Olivier Thibault
- Pharmacology and Nutritional Sciences University of Kentucky, University of Kentucky Medical Center, Lexington, KY, USA
| |
Collapse
|
5
|
Dludla PV, Nkambule BB, Mazibuko-Mbeje SE, Nyambuya TM, Silvestri S, Orlando P, Mxinwa V, Louw J, Tiano L. The impact of dimethyl sulfoxide on oxidative stress and cytotoxicity in various experimental models. Toxicology 2021. [DOI: 10.1016/b978-0-12-819092-0.00025-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
6
|
Chen H, Cross AC, Thakkar A, Xu H, Li A, Paull D, Noggle SA, Kruger L, Denton TT, Gibson GE. Selective linkage of mitochondrial enzymes to intracellular calcium stores differs between human-induced pluripotent stem cells, neural stem cells, and neurons. J Neurochem 2020; 156:867-879. [PMID: 32865230 DOI: 10.1111/jnc.15160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria and releasable endoplasmic reticulum (ER) calcium modulate neuronal calcium signaling, and both change in Alzheimer's disease (AD). The releasable calcium stores in the ER are exaggerated in fibroblasts from AD patients and in multiple models of AD. The activity of the alpha-ketoglutarate dehydrogenase complex (KGDHC), a key mitochondrial enzyme complex, is diminished in brains from AD patients, and can be plausibly linked to plaques and tangles. Our previous studies in cell lines and mouse neurons demonstrate that reductions in KGDHC increase the ER releasable calcium stores. The goal of these studies was to test whether the relationship was true in human iPSC-derived neurons. Inhibition of KGDHC for one or 24 hr increased the ER releasable calcium store in human neurons by 69% and 144%, respectively. The effect was mitochondrial enzyme specific because inhibiting the pyruvate dehydrogenase complex, another key mitochondrial enzyme complex, diminished the ER releasable calcium stores. The link of KGDHC to ER releasable calcium stores was cell type specific as the interaction was not present in iPSC or neural stem cells. Thus, these studies in human neurons verify a link between KGDHC and releasable ER calcium stores, and support the use of human neurons to examine mechanisms and potential therapies for AD.
Collapse
Affiliation(s)
- Huanlian Chen
- Burke Neurological Institute, Brain and Mind Research Institute, Weill Cornell Medicine, White Plains, NY, USA
| | - Abigail C Cross
- Burke Neurological Institute, Brain and Mind Research Institute, Weill Cornell Medicine, White Plains, NY, USA
| | - Ankita Thakkar
- Burke Neurological Institute, Brain and Mind Research Institute, Weill Cornell Medicine, White Plains, NY, USA
| | - Hui Xu
- Burke Neurological Institute, Brain and Mind Research Institute, Weill Cornell Medicine, White Plains, NY, USA
| | - Aiqun Li
- The New York Stem Cell Foundation Research Institute, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan Paull
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Scott A Noggle
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Laken Kruger
- Department of Pharmaceutical Sciences, Washington State University, College of Pharmacy and Pharmaceutical Sciences, Spokane, WA, USA
| | - Travis T Denton
- Department of Pharmaceutical Sciences, Washington State University, College of Pharmacy and Pharmaceutical Sciences, Spokane, WA, USA
| | - Gary E Gibson
- Burke Neurological Institute, Brain and Mind Research Institute, Weill Cornell Medicine, White Plains, NY, USA
| |
Collapse
|
7
|
Cystamine and cysteamine as inhibitors of transglutaminase activity in vivo. Biosci Rep 2018; 38:BSR20180691. [PMID: 30054429 PMCID: PMC6123069 DOI: 10.1042/bsr20180691] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/07/2018] [Accepted: 07/24/2018] [Indexed: 12/22/2022] Open
Abstract
Cystamine is commonly used as a transglutaminase inhibitor. This disulphide undergoes reduction in vivo to the aminothiol compound, cysteamine. Thus, the mechanism by which cystamine inhibits transglutaminase activity in vivo could be due to either cystamine or cysteamine, which depends on the local redox environment. Cystamine inactivates transglutaminases by promoting the oxidation of two vicinal cysteine residues on the enzyme to an allosteric disulphide, whereas cysteamine acts as a competitive inhibitor for transamidation reactions catalyzed by this enzyme. The latter mechanism is likely to result in the formation of a unique biomarker, N-(γ-glutamyl)cysteamine that could serve to indicate how cyst(e)amine acts to inhibit transglutaminases inside cells and the body.
Collapse
|
8
|
Chen H, Denton TT, Xu H, Calingasan N, Beal MF, Gibson GE. Reductions in the mitochondrial enzyme α-ketoglutarate dehydrogenase complex in neurodegenerative disease - beneficial or detrimental? J Neurochem 2017; 139:823-838. [PMID: 27580471 DOI: 10.1111/jnc.13836] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/09/2016] [Accepted: 08/19/2016] [Indexed: 01/10/2023]
Abstract
Reductions in metabolism and excess oxidative stress are prevalent in multiple neurodegenerative diseases. The activity of the mitochondrial enzyme α-ketoglutarate dehydrogenase complex (KGDHC) appears central to these abnormalities. KGDHC is diminished in multiple neurodegenerative diseases. KGDHC can not only be rate limiting for NADH production and for substrate level phosphorylation, but is also a source of reactive oxygen species (ROS). The goal of these studies was to determine how changes in KGDHC modify baseline ROS, the ability to buffer ROS, baseline glutathionylation, calcium modulation and cell death in response to external oxidants. In vivo, reducing KGDHC with adeno virus diminished neurogenesis and increased oxidative stress. In vitro, treatments of short duration increased ROS and glutathionylation and enhanced the ability of the cells to diminish the ROS from added oxidants. However, long-term reductions lessened the ability to diminish ROS, diminished glutathionylation and exaggerated oxidant-induced changes in calcium and cell death. Increasing KGDHC enhanced the ability of the cells to diminish externally added ROS and protected against oxidant-induced changes in calcium and cell death. The results suggest that brief periods of diminished KGDHC are protective, while prolonged reductions are harmful. Furthermore, elevated KGDHC activities are protective. Thus, mitogenic therapies that increase KGDHC may be beneficial in neurodegenerative diseases. Read the Editorial Highlight for this article on Page 689.
Collapse
Affiliation(s)
- Huanlian Chen
- Brain and Mind Research Institute, Weill Cornell Medical College, Burke Medical Research Institute, White Plains, New York, USA
| | - Travis T Denton
- Department of Pharmaceutical Sciences, Washington State University, College of Pharmacy, Spokane, Washington, USA
| | - Hui Xu
- Brain and Mind Research Institute, Weill Cornell Medical College, Burke Medical Research Institute, White Plains, New York, USA
| | - Noel Calingasan
- Brain and Mind Research Institute, Weill Cornell Medical College, York Avenue, New York, USA
| | - M Flint Beal
- Brain and Mind Research Institute, Weill Cornell Medical College, York Avenue, New York, USA
| | - Gary E Gibson
- Brain and Mind Research Institute, Weill Cornell Medical College, Burke Medical Research Institute, White Plains, New York, USA
| |
Collapse
|
9
|
Ribeiro ML, Moreira LM, Arçari DP, Dos Santos LF, Marques AC, Pedrazzoli J, Cerutti SM. Protective effects of chronic treatment with a standardized extract of Ginkgo biloba L. in the prefrontal cortex and dorsal hippocampus of middle-aged rats. Behav Brain Res 2016; 313:144-150. [PMID: 27424157 DOI: 10.1016/j.bbr.2016.06.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/10/2016] [Accepted: 06/14/2016] [Indexed: 12/25/2022]
Abstract
This study assessed the effects of chronic treatment with a standardized extract of Ginkgo biloba L. (EGb) on short-term and long-term memory as well as on anxiety-like and locomotor activity using the plus-maze discriminative avoidance task (PM-DAT). Additionally, we evaluated the antioxidant and neuroprotective effects of EGb on the prefrontal cortex (PFC) and dorsal hippocampus (DH) of middle-aged rats using the comet assay. Twelve-month-old male Wistar rats were administered vehicle or EGb (0.5mgkg(-1) or 1.0gkg(-1)) for 30days. Behavioural data showed that EGb treatment improved short-term memory. Neither an anti-anxiety effect nor a change in locomotor activity was observed. Twenty-four hours after the behavioural tests, the rats were decapitated, and the PFC and DH were quickly dissected out and prepared for the comet assay. The levels of DNA damage in the PFC were significantly lower in rats that were treated with 1.0gkg(-1) EGb. Both doses of EGb decreased H2O2-induced DNA breakage in cortical cells, whereas the levels of DNA damage in the EGb-treated animals were significantly lower than those in the control animals. No significant differences in the level of DNA damage in hippocampal cells were observed among the experimental groups. EGb treatment was not able to reduce H2O2-induced DNA damage in hippocampal cells. Altogether, our data provide the first demonstration that chronic EGb treatment improved the short-term memory of middle-aged rats, an effect that could be associated with a reduction in free radical production in the PFC. These data suggest that EGb treatment might increase the survival of cortical neurons and corroborate and extend the view that EGb has protective and therapeutic properties.
Collapse
Affiliation(s)
- Marcelo L Ribeiro
- Laboratório de Biologia Molecular-Unidade Integrada de Farmacologia e Gastroenterologia, Universidade São Francisco, Bragança Paulista, São Paulo, Brazil; Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de Sao Paulo, Diadema, São Paulo, Brazil
| | - Luciana M Moreira
- Laboratório de Biologia Molecular-Unidade Integrada de Farmacologia e Gastroenterologia, Universidade São Francisco, Bragança Paulista, São Paulo, Brazil; Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de Sao Paulo, Diadema, São Paulo, Brazil
| | - Demetrius P Arçari
- Laboratório de Biologia Molecular-Unidade Integrada de Farmacologia e Gastroenterologia, Universidade São Francisco, Bragança Paulista, São Paulo, Brazil; Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de Sao Paulo, Diadema, São Paulo, Brazil
| | - Letícia França Dos Santos
- Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de Sao Paulo, Diadema, São Paulo, Brazil; Universidade São Francisco, Bragança Paulista, São Paulo, Brazil
| | - Antônio Cezar Marques
- Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de Sao Paulo, Diadema, São Paulo, Brazil; Universidade São Francisco, Bragança Paulista, São Paulo, Brazil
| | - José Pedrazzoli
- Laboratório de Biologia Molecular-Unidade Integrada de Farmacologia e Gastroenterologia, Universidade São Francisco, Bragança Paulista, São Paulo, Brazil; Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de Sao Paulo, Diadema, São Paulo, Brazil
| | - Suzete M Cerutti
- Cellular and Behavioral Pharmacology Laboratory, Department of Biological Science, Universidade Federal de Sao Paulo, Diadema, São Paulo, Brazil; Universidade São Francisco, Bragança Paulista, São Paulo, Brazil.
| |
Collapse
|
10
|
Mohamed JS, Wilson JC, Myers MJ, Sisson KJ, Alway SE. Dysregulation of SIRT-1 in aging mice increases skeletal muscle fatigue by a PARP-1-dependent mechanism. Aging (Albany NY) 2015; 6:820-34. [PMID: 25361036 PMCID: PMC4247385 DOI: 10.18632/aging.100696] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Accumulation of reactive oxygen species (ROS) in skeletal muscles and the resulting decline in muscle performance are hallmarks of sarcopenia. However, the precise mechanism by which ROS results in a decline in muscle performance is unclear. We demonstrate that isometric-exercise concomitantly increases the activities of Silent information regulator 1 (SIRT-1) and Poly [ADP-ribose] polymerase (PARP-1), and that activated SIRT-1 physically binds with and inhibits PARP-1 activity by a deacetylation dependent mechanism in skeletal muscle from young mice. In contrast, skeletal muscle from aged mice displays higher PARP-1 activity and lower SIRT-1 activity due to decreased intracellular NAD+ content, and as a result reduced muscle performance in response to exercise. Interestingly, injection of PJ34, a PARP-1 inhibitor, in aged mice increased SIRT-1 activity by preserving intracellular NAD+ content, which resulted in higher skeletal muscle mitochondrial biogenesis and performance. We found that the higher activity of PARP-1 in H2O2-treated myotubes or in exercised-skeletal muscles from aged mice is due to an elevated level of PARP-1 acetylation by the histone acetyltransferase General control of amino acid synthesis protein 5-like 2 (GCN-5). These results suggest that activation of SIRT-1 and/or inhibition of PARP-1 may ameliorate skeletal muscle performance in pathophysiological conditions such as sarcopenia and disuse-induced atrophy in aging.
Collapse
Affiliation(s)
- Junaith S Mohamed
- Laboratory of Muscle Biology and Sarcopenia, Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA. Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA
| | - Joseph C Wilson
- Laboratory of Muscle Biology and Sarcopenia, Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA. Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA
| | - Matthew J Myers
- Laboratory of Muscle Biology and Sarcopenia, Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA. Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA
| | - Kayla J Sisson
- Laboratory of Muscle Biology and Sarcopenia, Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA
| | - Stephen E Alway
- Laboratory of Muscle Biology and Sarcopenia, Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA. Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227, USA. West Virginia Clinical and Translational Science Institute, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9227 USA
| |
Collapse
|
11
|
Poly(ADP-ribose) polymerase 1 promotes oxidative-stress-induced liver cell death via suppressing farnesoid X receptor α. Mol Cell Biol 2013; 33:4492-503. [PMID: 24043304 DOI: 10.1128/mcb.00160-13] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Farnesoid X receptor α (FXR) is highly expressed in the liver and regulates the expression of various genes involved in liver repair. In this study, we demonstrated that activated poly(ADP-ribose) polymerase 1 (PARP1) promoted hepatic cell death by inhibiting the expression of FXR-dependent hepatoprotective genes. PARP1 could bind to and poly(ADP-ribosyl)ate FXR. Poly(ADP-ribosyl)ation dissociated FXR from the FXR response element (FXRE), present in the promoters of target genes, and suppressed FXR-mediated gene transcription. Moreover, treatment with a FXR agonist attenuated poly(ADP-ribosyl)ation of FXR and promoted FXR-dependent gene expression. We further established the CCl4-induced acute liver injury model in wild-type and FXR-knockout mice and identified an essential role of FXR poly(ADP-ribosyl)ation in CCl4-induced liver injury. Thus, our results identified poly(ADP-ribosyl)ation of FXR by PARP1 as a key step in oxidative-stress-induced hepatic cell death. The molecular association between PARP1 and FXR provides new insight into the mechanism, suggesting that inhibition of PARP1 could prevent liver injury.
Collapse
|
12
|
Hardas SS, Sultana R, Clark AM, Beckett TL, Szweda LI, Murphy MP, Butterfield DA. Oxidative modification of lipoic acid by HNE in Alzheimer disease brain. Redox Biol 2013; 1:80-5. [PMID: 24024140 PMCID: PMC3757677 DOI: 10.1016/j.redox.2013.01.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 12/27/2012] [Accepted: 01/02/2013] [Indexed: 12/19/2022] Open
Abstract
Alzheimer disease (AD) is an age-related neurodegenerative disease characterized by the presence of three pathological hallmarks: synapse loss, extracellular senile plaques (SP) and intracellular neurofibrillary tangles (NFTs). The major component of SP is amyloid β-peptide (Aβ), which has been shown to induce oxidative stress. The AD brain shows increased levels of lipid peroxidation products, including 4-hydroxy-2-nonenal (HNE). HNE can react covalently with Cys, His, or Lys residues on proteins, altering structure and function of the latter. In the present study we measured the levels of the HNE-modified lipoic acid in brain of subjects with AD and age-matched controls. Lipoic acid is a key co-factor for a number of proteins including pyruvate dehydrogenase and α-ketoglutarate dehydrogenase, key complexes for cellular energetics. We observed a significant decrease in the levels of HNE-lipoic acid in the AD brain compared to that of age-matched controls. To investigate this phenomenon further, the levels and activity of lipoamide dehydrogenase (LADH) were measured in AD and control brains. Additionally, LADH activities were measured after in-vitro HNE-treatment to mice brains. Both LADH levels and activities were found to be significantly reduced in AD brain compared to age-matched control. HNE-treatment also reduced the LADH activity in mice brain. These data are consistent with a two-hit hypothesis of AD: oxidative stress leads to lipid peroxidation that, in turn, causes oxidative dysfunction of key energy-related complexes in mitochondria, triggering neurodegeneration. This study is consonant with the notion that lipoic acid supplementation could be a potential treatment for the observed loss of cellular energetics in AD and potentiate the antioxidant defense system to prevent or delay the oxidative stress in and progression of this devastating dementing disorder.
Collapse
Affiliation(s)
- Sarita S Hardas
- Department of Chemistry, University of Kentucky, Lexington, KY 40506-0055, USA ; Center for Membrane Sciences, University of Kentucky, Lexington, KY 40506-0055, USA ; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Gibson GE, Chen HL, Xu H, Qiu L, Xu Z, Denton TT, Shi Q. Deficits in the mitochondrial enzyme α-ketoglutarate dehydrogenase lead to Alzheimer's disease-like calcium dysregulation. Neurobiol Aging 2012; 33:1121.e13-24. [PMID: 22169199 PMCID: PMC3321099 DOI: 10.1016/j.neurobiolaging.2011.11.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 10/27/2011] [Accepted: 11/03/2011] [Indexed: 01/08/2023]
Abstract
Understanding the molecular sequence of events that culminate in multiple abnormalities in brains from patients that died with Alzheimer's disease (AD) will help to reveal the mechanisms of the disease and identify upstream events as therapeutic targets. The activity of the mitochondrial α-ketoglutarate dehydrogenase complex (KGDHC) in homogenates from autopsy brain declines with AD. Experimental reductions in KGDHC in mouse models of AD promote plaque and tangle formation, the hallmark pathologies of AD. We hypothesize that deficits in KGDHC also lead to the abnormalities in endoplasmic reticulum (ER) calcium stores and cytosolic calcium following K(+) depolarization that occurs in cells from AD patients and transgenic models of AD. The activity of the mitochondrial enzyme KGDHC was diminished acutely (minutes), long-term (days), or chronically (weeks). Acute inhibition of KGDHC produced effects on calcium opposite to those in AD, while the chronic or long-term inhibition of KGDHC mimicked the AD-related changes in calcium. Divergent changes in proteins released from the mitochondria that affect endoplasmic reticulum calcium channels may underlie the selective cellular consequences of acute versus longer term inhibition of KGDHC. The results suggest that the mitochondrial abnormalities in AD can be upstream of those in calcium.
Collapse
Affiliation(s)
- Gary E Gibson
- Department of Neurology and Neuroscience, Weill Cornell Medical College, Burke Medical Research Institute, White Plains, NY 10605, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Chakraborty R, Vepuri V, Mhatre SD, Paddock BE, Miller S, Michelson SJ, Delvadia R, Desai A, Vinokur M, Melicharek DJ, Utreja S, Khandelwal P, Ansaloni S, Goldstein LE, Moir RD, Lee JC, Tabb LP, Saunders AJ, Marenda DR. Characterization of a Drosophila Alzheimer's disease model: pharmacological rescue of cognitive defects. PLoS One 2011; 6:e20799. [PMID: 21673973 PMCID: PMC3108982 DOI: 10.1371/journal.pone.0020799] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 05/13/2011] [Indexed: 02/07/2023] Open
Abstract
Transgenic models of Alzheimer's disease (AD) have made significant contributions to our understanding of AD pathogenesis, and are useful tools in the development of potential therapeutics. The fruit fly, Drosophila melanogaster, provides a genetically tractable, powerful system to study the biochemical, genetic, environmental, and behavioral aspects of complex human diseases, including AD. In an effort to model AD, we over-expressed human APP and BACE genes in the Drosophila central nervous system. Biochemical, neuroanatomical, and behavioral analyses indicate that these flies exhibit aspects of clinical AD neuropathology and symptomology. These include the generation of Aβ(40) and Aβ(42), the presence of amyloid aggregates, dramatic neuroanatomical changes, defects in motor reflex behavior, and defects in memory. In addition, these flies exhibit external morphological abnormalities. Treatment with a γ-secretase inhibitor suppressed these phenotypes. Further, all of these phenotypes are present within the first few days of adult fly life. Taken together these data demonstrate that this transgenic AD model can serve as a powerful tool for the identification of AD therapeutic interventions.
Collapse
Affiliation(s)
- Ranjita Chakraborty
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Vidya Vepuri
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Siddhita D. Mhatre
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Brie E. Paddock
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Sean Miller
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Sarah J. Michelson
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Radha Delvadia
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Arkit Desai
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Marianna Vinokur
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - David J. Melicharek
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Suruchi Utreja
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Preeti Khandelwal
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Sara Ansaloni
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Lee E. Goldstein
- Department of Psychiatry, Boston University, Boston, Massachusetts, United States of America
| | - Robert D. Moir
- Genetics and Aging Research Unit, MIND, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jeremy C. Lee
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Loni P. Tabb
- Department of Epidemiology and Biostatistics, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Aleister J. Saunders
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Daniel R. Marenda
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, United States of America
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
15
|
Detection and manipulation of mitochondrial reactive oxygen species in mammalian cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1797:1034-44. [PMID: 20100455 DOI: 10.1016/j.bbabio.2010.01.022] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 01/13/2010] [Accepted: 01/18/2010] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are formed upon incomplete reduction of molecular oxygen (O2) as an inevitable consequence of mitochondrial metabolism. Because ROS can damage biomolecules, cells contain elaborate antioxidant defense systems to prevent oxidative stress. In addition to their damaging effect, ROS can also operate as intracellular signaling molecules. Given the fact that mitochondrial ROS appear to be only generated at specific sites and that particular ROS species display a unique chemistry and have specific molecular targets, mitochondria-derived ROS might constitute local regulatory signals. The latter would allow individual mitochondria to auto-regulate their metabolism, shape and motility, enabling them to respond autonomously to the metabolic requirements of the cell. In this review we first summarize how mitochondrial ROS can be generated and removed in the living cell. Then we discuss experimental strategies for (local) detection of ROS by combining chemical or proteinaceous reporter molecules with quantitative live cell microscopy. Finally, approaches involving targeted pro- and antioxidants are presented, which allow the local manipulation of ROS levels.
Collapse
|
16
|
Abstract
Alzheimer's disease (AD) is defined by senile plaques made of amyloid-beta peptide (Abeta), neurofibrillary tangles made of hyperphosphorylated tau proteins, and memory deficits. Thus, the events initiating the cascade leading to these end points may be more effective therapeutic targets than treating each facet individually. In the small percentage of cases of AD that are genetic (or animal models that reflect this form of AD), the factor initiating AD is clear (e.g., genetic mutations lead to high Abeta1-42 or hyperphosphorylated tau proteins). In the vast majority of AD cases, the cause is unknown. Substantial evidence now suggests that abnormalities in glucose metabolism/mitochondrial function/oxidative stress (GMO) are an invariant feature of AD and occur at an early stage of the disease process in both genetic and non-genetic forms of AD. Indeed, decreases in brain glucose utilization are diagnostic for AD. Changes in calcium homeostasis also precede clinical manifestations of AD. Abnormal GMO can lead to plaques, tangles, and the calcium abnormalities that accompany AD. Abnormalities in GMO diminish the ability of the brain to adapt. Therapies targeting mitochondria may ameliorate abnormalities in plaques, tangles, calcium homeostasis, and cognition that comprise AD.
Collapse
Affiliation(s)
- Gary E Gibson
- Weill Cornell Medical College/Burke Medical Research Institute, White Plains, NY, USA.
| | | |
Collapse
|
17
|
Gibson GE, Starkov A, Blass JP, Ratan RR, Beal MF. Cause and consequence: mitochondrial dysfunction initiates and propagates neuronal dysfunction, neuronal death and behavioral abnormalities in age-associated neurodegenerative diseases. Biochim Biophys Acta Mol Basis Dis 2009; 1802:122-34. [PMID: 19715758 DOI: 10.1016/j.bbadis.2009.08.010] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 08/14/2009] [Accepted: 08/17/2009] [Indexed: 12/31/2022]
Abstract
Age-related neurodegenerative diseases are associated with mild impairment of oxidative metabolism and accumulation of abnormal proteins. Within the cell, the mitochondria appears to be a dominant site for initiation and propagation of disease processes. Shifts in metabolism in response to mild metabolic perturbations may decrease the threshold for irreversible injury in response to ordinarily sublethal metabolic insults. Mild impairment of metabolism accrue from and lead to increased reactive oxygen species (ROS). Increased ROS change cell signaling via post-transcriptional and transcriptional changes. The cause and consequences of mild impairment of mitochondrial metabolism is one focus of this review. Many experiments in tissues from humans support the notion that oxidative modification of the alpha-ketoglutarate dehydrogenase complex (KGDHC) compromises neuronal energy metabolism and enhances ROS production in Alzheimer's Disease (AD). These data suggest that cognitive decline in AD derives from the selective tricarboxylic acid (TCA) cycle abnormalities. By contrast in Huntington's Disease (HD), a movement disorder with cognitive features distinct form AD, complex II+III abnormalities may dominate. These distinct mitochondrial abnormalities culminate in oxidative stress, energy dysfunction, and aberrant homeostasis of cytosolic calcium. Cytosolic calcium, elevations even only transiently, leads to hyperactivity of a number of enzymes. One calcium-activated enzyme with demonstrated pathophysiological import in HD and AD is transglutaminase (TGase). TGase is a crosslinking enzymes that can modulate transcription, inactivate metabolic enzymes, and cause aggregation of critical proteins. Recent data indicate that TGase can silence expression of genes involved in compensating for metabolic stress. Altogether, our results suggest that increasing KGDHC via inhibition of TGase or via a host of other strategies to be described would be effective therapeutic approaches in age-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Gary E Gibson
- Department of Neurology and Neuroscience, Weill Cornell Medical College of Cornell University at Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA.
| | | | | | | | | |
Collapse
|
18
|
Distelmaier F, Visch HJ, Smeitink JAM, Mayatepek E, Koopman WJH, Willems PHGM. The antioxidant Trolox restores mitochondrial membrane potential and Ca2+ -stimulated ATP production in human complex I deficiency. J Mol Med (Berl) 2009; 87:515-22. [PMID: 19255735 PMCID: PMC2836243 DOI: 10.1007/s00109-009-0452-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 01/22/2009] [Accepted: 02/10/2009] [Indexed: 11/29/2022]
Abstract
Malfunction of mitochondrial complex I caused by nuclear gene mutations causes early-onset neurodegenerative diseases. Previous work using cultured fibroblasts of complex-I-deficient patients revealed elevated levels of reactive oxygen species (ROS) and reductions in both total Ca(2+) content of the endoplasmic reticulum (ER(Ca)) and bradykinin(Bk)-induced increases in cytosolic and mitochondrial free Ca(2+) ([Ca(2+)](C); [Ca(2+)](M)) and ATP ([ATP](C); [ATP](M)) concentration. Here, we determined the mitochondrial membrane potential (Delta psi) in patient skin fibroblasts and show significant correlations with cellular ROS levels and ER(Ca), i.e., the less negative Delta psi, the higher these levels and the lower ER(Ca). Treatment with 6-hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid (Trolox) normalized Delta psi and Bk-induced increases in [Ca(2+)](M) and [ATP](M). These effects were accompanied by an increase in ER(Ca) and Bk-induced increase in [Ca(2+)](C). Together, these results provide evidence for an integral role of increased ROS levels in complex I deficiency and point to the potential therapeutic value of antioxidant treatment.
Collapse
Affiliation(s)
- Felix Distelmaier
- Department of Biochemistry (286), Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
19
|
Gibson GE, Karuppagounder SS, Shi Q. Oxidant-induced changes in mitochondria and calcium dynamics in the pathophysiology of Alzheimer's disease. Ann N Y Acad Sci 2008; 1147:221-32. [PMID: 19076444 PMCID: PMC2744687 DOI: 10.1196/annals.1427.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Considerable data support the hypothesis that mitochondrial abnormalities link gene defects and/or environmental insults to the neurodegenerative process. The interaction of oxidants with calcium and the mitochondrial enzymes of the tricarboxylic acid cycle are central to that relationship. Abnormalities that were discovered in brains or fibroblasts from patients with Alzheimer's disease (AD) have been modeled in vitro and in vivo to assess their pathophysiological importance and to determine how they might be reversed. The conclusions are consistent with the hypothesis that the AD-related abnormalities result from oxidative stress. The selection of compounds for reversal is complex because the actions of the relevant compounds vary under different conditions, such as cell redox states and acute versus chronic changes. However, the models that have been developed are useful for testing the effectiveness of the potential medications. The results suggest that the reversal of mitochondrial deficits and a reduction in oxidative stress will reduce clinical and pathological changes and benefit patients.
Collapse
Affiliation(s)
- Gary E Gibson
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, NY 10605, USA.
| | | | | |
Collapse
|
20
|
Shi Q, Xu H, Kleinman WA, Gibson GE. Novel functions of the alpha-ketoglutarate dehydrogenase complex may mediate diverse oxidant-induced changes in mitochondrial enzymes associated with Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1782:229-38. [PMID: 18206986 PMCID: PMC3106300 DOI: 10.1016/j.bbadis.2007.12.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 12/07/2007] [Accepted: 12/12/2007] [Indexed: 12/13/2022]
Abstract
Measures in autopsied brains from Alzheimer's Disease (AD) patients reveal a decrease in the activity of alpha-ketoglutarate dehydrogenase complex (KGDHC) and an increase in malate dehydrogenase (MDH) activity. The present experiments tested whether both changes could be caused by the common oxidant H(2)O(2) and to probe the mechanism underlying these changes. Since the response to H(2)O(2) is modified by the level of the E2k subunit of KGDHC, the interaction of MDH and KGDHC was studied in cells with varying levels of E2k. In cells with only 23% of normal E2k protein levels, one-hour treatment with H(2)O(2) decreased KGDHC and increased MDH activity as well as the mRNA level for both cytosolic and mitochondrial MDH. The increase in MDH did not occur in cells with 100% or 46% of normal E2k. Longer treatments with H(2)O(2) inhibited the activity of both enzymes. Glutathione is a major regulator of cellular redox state and can modify enzyme activities. H(2)O(2) converts reduced glutathione (GSH) to oxidized glutathione (GSSG), which reacts with protein thiols. Treatment of purified KGDHC with GSSG leads to glutathionylation of all three KGDHC subunits. Thus, cellular glutathione level was manipulated by two means to determine the effect on KGDHC and MDH activities. Both buthionine sulfoximine (BSO), which inhibits glutathione synthesis without altering redox state, and H(2)O(2) diminished glutathione to a similar level after 24 h. However, H(2)O(2), but not BSO, reduced KGDHC and MDH activities, and the reduction was greater in the E2k-23 line. These findings suggest that the E2k may mediate diverse responses of KGDHC and MDH to oxidants. In addition, the differential response of activities to BSO and H(2)O(2) together with the in vitro interaction of KGDHC with GSSG suggests that glutathionylation is one possible mechanism underlying oxidative stress-induced inhibition of the TCA cycle enzymes.
Collapse
Affiliation(s)
- Qingli Shi
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University/Burke Medical Research Institute, White Plains, New York 10605, USA
| | | | | | | |
Collapse
|
21
|
Abstract
Reductions in brain glucose metabolism and increased oxidative stress invariably occur in Alzheimer's disease (AD) and thiamine (vitamin B1) deficiency. Both conditions cause irreversible cognitive impairment; their behavioral consequences overlap but are not identical. Thiamine-dependent processes are critical in glucose metabolism, and recent studies implicate thiamine in oxidative stress, protein processing, peroxisomal function, and gene expression. The activities of thiamine-dependent enzymes are characteristically diminished in AD, and the reductions in autopsy AD brain correlate highly with the extent of dementia in the preagonal state. Abnormalities in thiamine-dependent processes can be plausibly linked to the pathology of AD. Seemingly paradoxical properties of thiamine-dependent processes may underlie their relation to the pathophysiology of AD: Reduction of thiamine-dependent processes increase oxidative stress. Thiamine can act as a free radical scavenger. Thiamine-dependent mitochondrial dehydrogenase complexes produce oxygen free radicals and are sensitive to oxidative stress. Genetic disorders of thiamine metabolism that lead to neurological disease can be treated with large doses of thiamine. Although thiamine itself has not shown dramatic benefits in AD patients, the available data is scanty. Adding thiamine or more absorbable forms of thiamine to tested treatments for the abnormality in glucose metabolism in AD may increase their efficacy.
Collapse
Affiliation(s)
- Gary E Gibson
- Department of Neurology and Neurosciences, Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, New York 10605, USA.
| | | |
Collapse
|
22
|
Abstract
Alzheimer disease (AD) is defined by progressive impairments in memory and cognition and by the presence of extracellular neuritic plaques and intracellular neurofibrillary tangles. However, oxidative stress and impaired mitochondrial function always accompany AD. Mitochondria are a major site of production of free radicals [ie, reactive oxygen species (ROS)] and primary targets of ROS. ROS are cytotoxic, and evidence of ROS-induced damage to cell membranes, proteins, and DNA in AD is overwhelming. Nevertheless, therapies based on antioxidants have been disappointing. Thus, alternative strategies are necessary. ROS also act as signaling molecules including for transcription. Thus, chronic exposure to ROS in AD could activate cascades of genes. Although initially protective, prolonged activation may be damaging. Thus, therapeutic approaches based on modulation of these gene cascades may lead to effective therapies. Genes involved in several pathways including antioxidant defense, detoxification, inflammation, etc, are induced in response to oxidative stress and in AD. However, genes that are associated with energy metabolism, which is necessary for normal brain function, are mostly down-regulated. Redox-sensitive transcription factors such as activator protein-1, nuclear factor-kappaB, specificity protein-1, and hypoxia-inducible factor are important in redox-dependent gene regulation. Peroxisome proliferators-activated receptor-gamma coactivator (PGC-1alpha) is a coactivator of several transcription factors and is a potent stimulator of mitochondrial biogenesis and respiration. Down-regulated expression of PGC-1alpha has been implicated in Huntington disease and in several Huntington disease animal models. PGC-1alpha role in regulation of ROS metabolism makes it a potential candidate player between ROS, mitochondria, and neurodegenerative diseases. This review summarizes the current progress on how oxidative stress regulates the expression of genes that might contribute to AD pathophysiology and the implications of the transcriptional modifications for AD. Finally, potential therapeutic strategies based on the updated understandings of redox state-dependent gene regulation in AD are proposed to overcome the lack of efficacy of antioxidant therapies.
Collapse
Affiliation(s)
- Qingli Shi
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, NY 10605, USA.
| | | |
Collapse
|
23
|
Butterfield DA, Reed T, Newman SF, Sultana R. Roles of amyloid beta-peptide-associated oxidative stress and brain protein modifications in the pathogenesis of Alzheimer's disease and mild cognitive impairment. Free Radic Biol Med 2007; 43:658-77. [PMID: 17664130 PMCID: PMC2031860 DOI: 10.1016/j.freeradbiomed.2007.05.037] [Citation(s) in RCA: 440] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2007] [Revised: 05/20/2007] [Accepted: 05/25/2007] [Indexed: 12/17/2022]
Abstract
Oxidative stress has been implicated to play a crucial role in the pathogenesis of a number of diseases, including neurodegenerative disorders, cancer, and ischemia, just to name a few. Alzheimer disease (AD) is an age-related neurodegenerative disorder that is recognized as the most common form of dementia. AD is histopathologically characterized by the presence of extracellular amyloid plaques, intracellular neurofibrillary tangles, the presence of oligomers of amyloid beta-peptide (Abeta), and synapse loss. In this review we discuss the role of Abeta in the pathogenesis of AD and also the use of redox proteomics to identify oxidatively modified brain proteins in AD and mild cognitive impairment. In addition, redox proteomics studies in in vivo models of AD centered around human Abeta(1-42) are discussed.
Collapse
|
24
|
Shi Q, Karuppagounder SS, Xu H, Pechman D, Chen H, Gibson GE. Responses of the mitochondrial alpha-ketoglutarate dehydrogenase complex to thiamine deficiency may contribute to regional selective vulnerability. Neurochem Int 2007; 50:921-31. [PMID: 17482317 PMCID: PMC2753422 DOI: 10.1016/j.neuint.2007.03.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 03/15/2007] [Accepted: 03/26/2007] [Indexed: 11/22/2022]
Abstract
Thiamine-dependent enzymes are diminished in multiple neurodegenerative diseases. Thiamine deficiency (TD) reduces the activity of thiamine dependent-enzymes [e.g., the alpha-ketoglutarate dehydrogenase complex (KGDHC)], induces regional selective neurodegeneration and serves as a model of a mild impairment of oxidative metabolism. The current experiments tested whether changes in KGDHC protein subunits (E1k, E2k and E3) or activity or message levels underlie the selective loss of neurons in particular brain regions. Thus, TD-induced changes in these variables in the brain region most vulnerable to TD [the sub-medial thalamic nucleus (SmTN)] were compared to those in a region that is relatively resistant to TD (cortex) at stages of TD when the neuron loss in SmTN is not present, minimal or severe. Impaired motor performance on rotarod was apparent by 8 days of TD (-32%) and was severe by 10 days of TD (-97%). At TD10, the overall KGDHC activity measured by an in situ histochemical staining method declined 52% in SmTN but only 20% in cortex. Reductions in the E2k and E3 mRNA in SmTN occurred as early as TD6 (-28 and -18%, respectively) and were more severe by TD10 (-61 and -66%, respectively). On the other hand, the level of E1k mRNA did not decline in SmTN until TD10 (-48%). In contrast, TD did not alter mRNA levels of the subunits in cortex at late stages. Western blots and immunocytochemistry revealed different aspects of the changes in protein levels. In SmTN, the immunoreactivity of E1k and E3 by Western blotting increased 34 and 40%, respectively, only at TD8. In cortex, the immunoreactivity of the three subunits was not altered. Immunocytochemical staining of brain sections from TD10 mice indicated a reduction in the immunoreactivity of all subunits in SmTN, but not in cortex. These findings demonstrate that the response of the KGDHC activity, mRNA and immunoreactivity of E1k, E2k and E3 to TD is region and time dependent. Loss of KGDHC activity in cortex is likely related to post-translational modification rather than a loss of protein, whereas in SmTN transcriptional and post-translational modifications may account for diminished KGDHC activity. Moreover, the earlier detection in TD induced-changes of the transcripts of KGDHC indicates that transcriptional modification of the two subunits (E2k and E3) of KGDHC may be one of the early events in the cascade leading to selective neuronal death.
Collapse
Affiliation(s)
| | | | - H. Xu
- Dept of Neurology and Neuroscience, Weill Medical College of Cornell University at Burke Medical Research Institute, White Plains, NY 10605
| | - D. Pechman
- Dept of Neurology and Neuroscience, Weill Medical College of Cornell University at Burke Medical Research Institute, White Plains, NY 10605
| | - H. Chen
- Dept of Neurology and Neuroscience, Weill Medical College of Cornell University at Burke Medical Research Institute, White Plains, NY 10605
| | - G. E. Gibson
- Dept of Neurology and Neuroscience, Weill Medical College of Cornell University at Burke Medical Research Institute, White Plains, NY 10605
| |
Collapse
|
25
|
Wang X, Dykens JA, Perez E, Liu R, Yang S, Covey DF, Simpkins JW. Neuroprotective effects of 17beta-estradiol and nonfeminizing estrogens against H2O2 toxicity in human neuroblastoma SK-N-SH cells. Mol Pharmacol 2006; 70:395-404. [PMID: 16614138 DOI: 10.1124/mol.106.022384] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuroprotective effects of estrogens have been shown in various in vitro and in vivo models, but the mechanisms underlying protection by estrogen are not clear. Mounting evidence suggests antioxidant effects contribute to the neuroprotective effects of estrogens. In the present study, we assessed the protective effects of estrogens against H2O2-induced toxicity in human neuroblastoma cells and the potential mechanisms involved in this protection. We demonstrate that 17beta-estradiol (17beta-E2) increases cell survival against H2O2 toxicity in human neuroblastoma cells. 17beta-E2 effectively reduced lipid peroxidation induced by 5-min H2O2 exposure. Furthermore, 17beta-E2 exerts the protective effects by maintaining intracellular Ca2+ homeostasis, attenuating ATP depletion, ablating mitochondrial calcium overloading, and preserving mitochondrial membrane potential. Two nonfeminizing estrogens, 17alpha- and ent-estradiol, were as effective as 17beta-E2 in increasing cell survival, alleviating lipid peroxidation, preserving mitochondrial function, and maintaining intracellular glutathione levels and Ca2+ homeostasis against H2O2 insult. Moreover, the estrogen receptor antagonist fulvestrant (ICI 182,780) did not block effects of 17beta-E2, but increased cell survival and blunted intracellular Ca2+ increases. However, these estrogens failed to reduce cytosolic reactive oxygen species, even at concentrations as high as 10 microM. In conclusion, estrogens exert protective effects against oxidative stress by inhibiting lipid peroxidation and subsequently preserving Ca2+ homeostasis, mitochondrial membrane potential, and ATP levels.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Huang HM, Chen HL, Xu H, Gibson GE. Modification of endoplasmic reticulum Ca2+ stores by select oxidants produces changes reminiscent of those in cells from patients with Alzheimer disease. Free Radic Biol Med 2005; 39:979-89. [PMID: 16198225 DOI: 10.1016/j.freeradbiomed.2005.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2005] [Revised: 04/19/2005] [Accepted: 05/11/2005] [Indexed: 11/28/2022]
Abstract
Abnormalities in calcium homeostasis and oxidative processes occur in fibroblasts from patients with Alzheimer disease (AD) and in fibroblasts and neurons from transgenic mice bearing a presenilin-1 (PS-1) mutation. Bombesin-releasable endoplasmic reticulum Ca2+ stores (BRCS) are exaggerated in all of these cells. Our previous studies show that H2O2 exaggerates BRCS. The goal of the present study was to determine whether select reactive species exaggerate BRCS in cultured human fibroblasts and to determine if the ability of fibroblasts to handle these specific oxidant species is altered in cells from AD patients. Two fluorescent indicators were used to distinguish different reactive oxygen species (ROS): 6-carboxy-2',7'-dichlorodihydrofluorescein diacetate, di(acetoxymethyl ester) (c-DCF) and 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM). ROS were produced by a variety of oxidants, including tert-butyl-hydroxyperoxide (t-BHP), hypoxanthine/xanthine oxidase, S-nitroso-N-acetylpenicillamine, 3-morpholinosydnonimine, and sodium nitroprusside. Different oxidants selectively induced various ROS in distinct patterns. These oxidants also induced selective modification in [Ca2+]i and/or BRCS. Of the several oxidants tested, t-BHP was most specific for exaggerating BRCS without affecting basal [Ca2+]i and inducing only c-DCF-detectable ROS. On the other hand, the results show that NO that reacted with DAF-FM was not responsible for alterations in BRCS. Furthermore, the c-DCF-detectable ROS production induced by t-BHP was higher in fibroblasts from AD patients bearing a PS-1 mutation (n = 7) than in those from aged controls (n = 8). The higher production of c-DCF-detectable ROS may underlie the exaggeration of BRCS in fibroblasts from AD patients. Thus, these results are consistent with the hypothesis that abnormalities in selective cellular ROS cause AD-related changes in intracellular calcium regulation.
Collapse
Affiliation(s)
- Hsueh-Meei Huang
- Weill Medical College of Cornell University at Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA.
| | | | | | | |
Collapse
|
27
|
Jeitner TM, Xu H, Gibson GE. Inhibition of the alpha-ketoglutarate dehydrogenase complex by the myeloperoxidase products, hypochlorous acid and mono-N-chloramine. J Neurochem 2005; 92:302-10. [PMID: 15663478 DOI: 10.1111/j.1471-4159.2004.02868.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Abstract alpha-Ketoglutarate dehydrogenase (KGDHC) complex activity is diminished in a number of neurodegenerative disorders and its diminution in Alzheimer Disease (AD) is thought to contribute to the major loss of cerebral energy metabolism that accompanies this disease. The loss of KGDHC activity appears to be predominantly due to post-translation modifications. Thiamine deficiency also results in decreased KGDHC activity and a selective neuronal loss. Recently, myeloperoxidase has been identified in the activated microglia of brains from AD patients and thiamine-deficient animals. Myeloperoxidase produces a powerful oxidant, hypochlorous acid that reacts with amines to form chloramines. The aim of this study was to investigate the ability of hypochlorous acid and chloramines to inhibit the activity of KGDHC activity as a first step towards investigating the role of myeloperoxidase in AD. Hypochlorous acid and mono-N-chloramine both inhibited purified and cellular KGDHC and the order of inhibition of the purified complex was hypochlorous acid (1x) > mono-N-chloramine (approximately 50x) > hydrogen peroxide (approximately 1,500). The inhibition of cellular KGDHC occurred with no significant loss of cellular viability at all exposure times that were examined. Thus, hypochlorous acid and chloramines have the potential to inactivate a major target in neurodegeneration.
Collapse
Affiliation(s)
- Thomas M Jeitner
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | |
Collapse
|
28
|
Gibson GE, Huang HM. Oxidative stress in Alzheimer's disease. Neurobiol Aging 2005; 26:575-8. [PMID: 15708429 DOI: 10.1016/j.neurobiolaging.2004.07.017] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2004] [Accepted: 07/09/2004] [Indexed: 12/01/2022]
Affiliation(s)
- Gary E Gibson
- Weill Medical College of Cornell University, Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA.
| | | |
Collapse
|
29
|
Migliore L, Fontana I, Colognato R, Coppede F, Siciliano G, Murri L. Searching for the role and the most suitable biomarkers of oxidative stress in Alzheimer's disease and in other neurodegenerative diseases. Neurobiol Aging 2005; 26:587-95. [PMID: 15708433 DOI: 10.1016/j.neurobiolaging.2004.10.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2004] [Accepted: 10/29/2004] [Indexed: 01/15/2023]
Abstract
The contribution of oxidative stress to neurodegeneration is not peculiar of a specific neurodegenerative disease, oxidative stress has been found implicated in a number of neurodegenerative disorders among which Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS). Even increasing are studies dealing with the search for peripheral biomarkers of oxidative stress in biological fluids or even in peripheral tissues themselves such as fibroblasts or blood cells. The application of the modified version of the comet assay for the detection of oxidised purines and pyrimidines in peripheral blood leukocytes results particularly useful if the study requires repeated blood drawn from the same individual, for instance if a clinical trial is performed with a preventive therapy. Likely damage occurs to every category of biological macromolecules and we consider, in the context of neurodegenerative diseases, particularly critical the proteic level. The identification of subjects at risk to develop AD or with pre-pathogenic conditions, the possibility to use "a battery of assays" for the detection of oxidative damage at peripheral level, together with recent advances in brain imaging, will allow to better address studies aimed not only to therapeutic purposes but also mainly to primary prevention.
Collapse
Affiliation(s)
- L Migliore
- Department of Human and Environmental Sciences, University of Pisa, Via S. Giuseppe 22, 56126 Pisa, Italy.
| | | | | | | | | | | |
Collapse
|
30
|
Gibson GE, Huang HM. Mitochondrial enzymes and endoplasmic reticulum calcium stores as targets of oxidative stress in neurodegenerative diseases. J Bioenerg Biomembr 2005; 36:335-40. [PMID: 15377868 DOI: 10.1023/b:jobb.0000041764.45552.f3] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Considerable evidence indicates that oxidative stress accompanies age-related neurodegenerative diseases. Specific mechanisms by which oxidative stress leads to neurodegeneration are unknown. Two targets of oxidative stress that are known to change in neurodegenerative diseases are the mitochondrial enzyme alpha-ketoglutarate dehydrogenase complex (KGDHC) and endoplasmic reticulum calcium stores. KGDHC activities are diminished in all common neurodegenerative diseases and the changes are particularly well documented in Alzheimer's disease (AD). A second change that occurs in cells from AD patients is an exaggerated endoplasmic reticulum calcium store [i.e., bombesin-releasable calcium stores (BRCS)]. H(2)O(2), a general oxidant, changes both variables in the same direction as occurs in disease. Other oxidants selectively alter these variables. Various antioxidants were used to help define the critical oxidant species that modifies these responses. All of the antioxidants diminish the oxidant-induced carboxy-dichlorofluorescein (cDCF) detectable reactive oxygen species (ROS), but have diverse actions on these cellular processes. For example, alpha-keto-beta-methyl-n-valeric acid (KMV) diminishes the H(2)O(2) effects on BRCS, while trolox and DMSO exaggerate the response. Acute trolox treatment does not alter H(2)O(2)-induced changes in KGDHC, whereas chronic treatment with trolox increases KGDHC almost threefold. The results suggest that KGDHC and BRCS provide targets by which oxidative stress may induce neurodegeneration and a useful tool for selecting antioxidants for reversing age-related neurodegeneration.
Collapse
Affiliation(s)
- Gary E Gibson
- Burke Medical Research Institute, Weill Medical College of Cornell University, 785 Mamaroneck Avenue, White Plains, New York 10605, USA
| | | |
Collapse
|
31
|
Starkov AA, Fiskum G, Chinopoulos C, Lorenzo BJ, Browne SE, Patel MS, Beal MF. Mitochondrial alpha-ketoglutarate dehydrogenase complex generates reactive oxygen species. J Neurosci 2005; 24:7779-88. [PMID: 15356189 PMCID: PMC6729932 DOI: 10.1523/jneurosci.1899-04.2004] [Citation(s) in RCA: 531] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mitochondria-produced reactive oxygen species (ROS) are thought to contribute to cell death caused by a multitude of pathological conditions. The molecular sites of mitochondrial ROS production are not well established but are generally thought to be located in complex I and complex III of the electron transport chain. We measured H(2)O(2) production, respiration, and NADPH reduction level in rat brain mitochondria oxidizing a variety of respiratory substrates. Under conditions of maximum respiration induced with either ADP or carbonyl cyanide p-trifluoromethoxyphenylhydrazone,alpha-ketoglutarate supported the highest rate of H(2)O(2) production. In the absence of ADP or in the presence of rotenone, H(2)O(2) production rates correlated with the reduction level of mitochondrial NADPH with various substrates, with the exception of alpha-ketoglutarate. Isolated mitochondrial alpha-ketoglutarate dehydrogenase (KGDHC) and pyruvate dehydrogenase (PDHC) complexes produced superoxide and H(2)O(2). NAD(+) inhibited ROS production by the isolated enzymes and by permeabilized mitochondria. We also measured H(2)O(2) production by brain mitochondria isolated from heterozygous knock-out mice deficient in dihydrolipoyl dehydrogenase (Dld). Although this enzyme is a part of both KGDHC and PDHC, there was greater impairment of KGDHC activity in Dld-deficient mitochondria. These mitochondria also produced significantly less H(2)O(2) than mitochondria isolated from their littermate wild-type mice. The data strongly indicate that KGDHC is a primary site of ROS production in normally functioning mitochondria.
Collapse
Affiliation(s)
- Anatoly A Starkov
- Department of Neurology and Neuroscience, Weill Medical College, Cornell University, New York, New York 10021, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Lee J, Kim YS, Choi DH, Bang MS, Han TR, Joh TH, Kim SY. Transglutaminase 2 Induces Nuclear Factor-κB Activation via a Novel Pathway in BV-2 Microglia. J Biol Chem 2004; 279:53725-35. [PMID: 15471861 DOI: 10.1074/jbc.m407627200] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transglutaminase 2 (TGase 2) expression is increased in inflammatory diseases. We demonstrated previously that inhibitors of TGase 2 reduce nitric oxide (NO) generation in a lipopolysaccharide (LPS)-treated microglial cell line. However, the precise mechanism by which TGase 2 promotes inflammation remains unclear. We found that TGase 2 activates the transcriptional activator nuclear factor (NF)-kappaB and thereby enhances LPS-induced expression of inducible nitric-oxide synthase. TGase 2 activates NF-kappaB via a novel pathway. Rather than stimulating phosphorylation and degradation of the inhibitory subunit alpha of NF-kappaB (I-kappaBalpha), TGase2 induces its polymerization. This polymerization results in dissociation of NF-kappaB and its translocation to the nucleus, where it is capable of up-regulating a host of inflammatory genes, including inducible nitric-oxide synthase and tumor necrosis factor alpha (TNF-alpha). Indeed, TGase inhibitors prevent depletion of monomeric I-kappaBalpha in the cytosol of cells overexpressing TGase 2. In an LPS-induced rat brain injury model, TGase inhibitors significantly reduced TNF-alpha synthesis. The findings are consistent with a model in which LPS-induced NF-kappaB activation is the result of phosphorylation of I-kappaBalpha by I-kappaB kinase as well as I-kappaBalpha polymerization by TGase 2. Safe and stable TGase2 inhibitors may be effective agents in diseases associated with inflammation.
Collapse
Affiliation(s)
- Jongmin Lee
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University and Burke Medical Research Institute, White Plains, NY 10605, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Huang HM, Zhang H, Ou HC, Chen HL, Gibson GE. alpha-keto-beta-methyl-n-valeric acid diminishes reactive oxygen species and alters endoplasmic reticulum Ca(2+) stores. Free Radic Biol Med 2004; 37:1779-89. [PMID: 15528037 DOI: 10.1016/j.freeradbiomed.2004.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Revised: 07/08/2004] [Accepted: 08/05/2004] [Indexed: 11/22/2022]
Abstract
Mitochondrial dysfunction and oxidative stress occur in neurodegenerative diseases. Other results show that bombesin-releasable calcium stores (BRCS) from the endoplasmic reticulum (ER) are exaggerated in fibroblasts from patients with Alzheimer's disease (AD) compared with controls and in fibroblasts from a young control treated with H(2)O(2). We hypothesize that alterations in oxidative stress underlie the exaggeration in BRCS in AD, and that appropriate antioxidants may be useful in treating this abnormality. Two indicators of different oxidant species were used to determine the effects of select oxidants on cellular oxidation status: carboxydichlorofluorescein (c-DCF) to detect reactive oxygen species (ROS), and 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF) to detect nitric oxide (NO(.-)). Various conditions that induce ROS, including H(2)O(2), oxygen/glucose deprivation, and 3-morpholinosyndnonimine (SIN-1), were used to test the ability of alpha-keto-ss-methyl-n-valeric acid (KMV) to scavenge ROS. KMV diminished c-DCF-detectable ROS that were induced by H(2)O(2), oxygen/glucose deprivation, or SIN-1 in PC12 cells, primary neuronal cultures, or fibroblasts. Furthermore, KMV reduced the H(2)O(2)-induced increase in BRCS and diminished the elevation in BRCS in cells from AD patients to control levels. On the other hand, DAF-detectable NO(.-) induced by SIN-1 was not scavenged by KMV and did not exaggerate BRCS. The results indicate that KMV is an effective antioxidant of c-DCF-detectable ROS. The effects of KMV are not cell type specific, but are ROS specific. The same H(2)O(2)-induced ROS that reacts with KMV may also underlie the changes in BRCS related to AD. Thus, KMV ameliorates the effects of ROS on calcium homeostasis related to oxidative stress and to AD.
Collapse
Affiliation(s)
- Hsueh-Meei Huang
- Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, NY 10605, USA.
| | | | | | | | | |
Collapse
|
34
|
Klivenyi P, Starkov AA, Calingasan NY, Gardian G, Browne SE, Yang L, Bubber P, Gibson GE, Patel MS, Beal MF. Mice deficient in dihydrolipoamide dehydrogenase show increased vulnerability to MPTP, malonate and 3-nitropropionic acid neurotoxicity. J Neurochem 2004; 88:1352-60. [PMID: 15009635 DOI: 10.1046/j.1471-4159.2003.02263.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Altered energy metabolism, including reductions in activities of the key mitochondrial enzymes alpha-ketoglutarate dehydrogenase complex (KGDHC) and pyruvate dehydrogenase complex (PDHC), are characteristic of many neurodegenerative disorders including Alzheimer's Disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Dihydrolipoamide dehydrogenase is a critical subunit of KGDHC and PDHC. We tested whether mice that are deficient in dihydrolipoamide dehydrogenase (Dld+/-) show increased vulnerability to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), malonate and 3-nitropropionic acid (3-NP), which have been proposed for use in models of PD and HD. Administration of MPTP resulted in significantly greater depletion of tyrosine hydroxylase-positive neurons in the substantia nigra of Dld+/- mice than that seen in wild-type littermate controls. Striatal lesion volumes produced by malonate and 3-NP were significantly increased in Dld+/- mice. Studies of isolated brain mitochondria treated with 3-NP showed that both succinate-supported respiration and membrane potential were suppressed to a greater extent in Dld+/- mice. KGDHC activity was also found to be reduced in putamen from patients with HD. These findings provide further evidence that mitochondrial defects may contribute to the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter Klivenyi
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York Presbyterian Hospital, New York 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The present paper demonstrates a remarkable pervasiveness of underlying Ca(2+) signaling motifs among the available biochemical findings in schizophrenic patients and among the major molecular hypotheses of this disease. In addition, the paper reviews the findings suggesting that Ca(2+) is capable of inducing structural and cognitive deficits seen in schizophrenia. The evidence of the ability of antipsychotic drugs to affect Ca(2+) signaling is also presented. Based on these data, it is proposed that altered Ca(2+) signaling may constitute the central unifying molecular pathology in schizophrenia. According to this hypothesis schizophrenia can result from alterations in multiple proteins and other molecules as long as these alterations lead to abnormalities in certain key aspects of intracellular Ca(2+) signaling cascades.
Collapse
Affiliation(s)
- Michael S Lidow
- Department of Biomedical Sciences and Program of Neuroscience, University of Maryland, Room 5-A-12, HHH, 666 W. Baltimore Street, Baltimore, MD 21201, USA.
| |
Collapse
|
36
|
Manea A, Constantinescu E, Popov D, Raicu M. Changes in oxidative balance in rat pericytes exposed to diabetic conditions. J Cell Mol Med 2004; 8:117-26. [PMID: 15090267 PMCID: PMC6740059 DOI: 10.1111/j.1582-4934.2004.tb00266.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Recent data indicate that the oxidative stress plays an important role in the pathogenesis of diabetes and its complications such as retinopathy, nephropathy and accelerated atherosclerosis. In diabetic retinopathy, it was demonstrated a selective loss of pericytes accompanied by capillary basement membrane thickening, increased permeability and neovascularization. This study was designed to investigate the role of diabetic conditions such as high glucose, AGE-Lysine, and angiotensin II in the modulation of antioxidant enzymes activities, glutathione level and reactive oxygen species (ROS) production in pericytes. The activity of antioxidant enzymes: superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) and total glutathione (GSH) was measured spectrophotometrically. The production of ROS was detected by spectrofluorimetry and fluorescence microscopy after loading the cells with 2'-7' dichlorofluoresceine diacetate; as positive control H2O2 was used. Intracellular calcium was determined using Fura 2 AM assay. The results showed that the cells cultured in high glucose alone, do not exhibit major changes in the antioxidant enzyme activities. The presence of AGE-Lys or Ang II induced the increase of SOD activity. Their combination decreased significantly GPx activity and GSH level. A three times increase in ROS production and a significant impairment of intracellular calcium homeostasis was detected in cells cultured in the presence of the three pro-diabetic agents used. In conclusion, our data indicate that diabetic conditions induce in pericytes: (i) an increase of ROS and SOD activity, (ii) a decrease in GPx activity and GSH level, (iii) a major perturbation of the intracellular calcium homeostasis. The data may explain the structural and functional abnormalities of pericytes characteristic for diabetic retinopathy.
Collapse
Affiliation(s)
- A Manea
- Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| | | | | | | |
Collapse
|
37
|
Huang HM, Ou HC, Xu H, Chen HL, Fowler C, Gibson GE. Inhibition of alpha-ketoglutarate dehydrogenase complex promotes cytochrome c release from mitochondria, caspase-3 activation, and necrotic cell death. J Neurosci Res 2003; 74:309-17. [PMID: 14515360 DOI: 10.1002/jnr.10756] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mitochondrial dysfunction has been implicated in cell death in many neurodegenerative diseases. Diminished activity of the alpha-ketoglutarate dehydrogenase complex (KGDHC), a key and arguably rate-limiting enzyme of the Krebs cycle, occurs in these disorders and may underlie decreased brain metabolism. The present studies used alpha-keto-beta-methyl-n-valeric acid (KMV), a structural analogue of alpha-ketoglutarate, to inhibit KGDHC activity to test effects of reduced KGDHC on mitochondrial function and cell death cascades in PC12 cells. KMV decreased in situ KGDHC activity by 52 +/- 7% (1 hr) or 65 +/- 4% (2 hr). Under the same conditions, KMV did not alter the mitochondrial membrane potential (MMP), as assessed with a method that detects changes as small as 5%. KMV also did not alter production of reactive oxygen species (ROS). However, KMV increased lactate dehydrogenase (LDH) release from cells by 100 +/- 4.7%, promoted translocation of mitochondrial cytochrome c to the cytosol, and activated caspase-3. Inhibition of the mitochondrial permeability transition pore (MPTP) by cyclosporin A (CsA) partially blocked this KMV-induced change in cytochrome c (-40%) and LDH (-15%) release, and prevented necrotic cell death. Thus, impairment of this key mitochondrial enzyme in PC12 cells may lead to cytochrome c release and caspase-3 activation by partial opening of the MPTP before the loss of mitochondrial membrane potentials.
Collapse
Affiliation(s)
- Hsueh-Meei Huang
- Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, New York 10605, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
Extensive, replicated evidence in patients in vivo and in Alzheimer (AD) tissues in vitro indicates that impaired brain metabolism is one of the cardinal and essentially invariable events in AD. The degree of impairment in brain metabolism is proportional to the degree of clinical disability, both in vivo and in vitro. The 'cerebrometabolic lesion' cannot be attributed to 'slower thinking' or 'brain atrophy', because of quantitative considerations and because the metabolic lesion precedes the development of neuropsychological abnormalities or decreases in brain mass detectable by modern imaging techniques. The causes of the cerebrometabolic lesion in AD are not well defined. Free radicals seem likely to be involved, including free radicals generated from Alzheimer amyloid. Thus, the importance of the cerebrometabolic lesion is entirely compatible with most versions of the widely accepted 'amyloid cascade hypothesis' of AD. A variety of plausible, redundantly documented mechanisms are compatible with the proposal that the cerebrometabolic lesion is a proximate cause of the clinical disability in AD. In agreement with these findings, recent attempts to treat the cerebrometabolic lesion in AD have given encouraging preliminary results. The cerebrometabolic lesion in AD deserves further study.
Collapse
Affiliation(s)
- John P Blass
- Dementia Research Service, Burke Medical Research Institute, Weill Medical College of Cornell University, 785 Mamaroneck Avenue, White Plains, NY 10708, USA.
| |
Collapse
|
39
|
Kim DK, Natarajan N, Prabhakar NR, Kumar GK. Facilitation of dopamine and acetylcholine release by intermittent hypoxia in PC12 cells: involvement of calcium and reactive oxygen species. J Appl Physiol (1985) 2003; 96:1206-15; discussion 1196. [PMID: 14657041 DOI: 10.1152/japplphysiol.00879.2003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have investigated the effects of preconditioning pheochromocytoma (PC12) cells with intermittent hypoxia (IH) on transmitter release during acute hypoxia. Cell cultures were exposed to either alternating cycles of hypoxia (1% O(2) + 5% CO(2); 30 s/cycle) and normoxia (21% O(2) + 5% CO(2); 3 min/cycle) for 15 or 60 cycles or normoxia alone (control) for similar durations. Control and IH cells were challenged with either hyperoxia (basal release) or acute hypoxia (Po(2) of approximately 35 Torr) for 5 min, and the amounts of dopamine (DA) and acetylcholine (ACh) released in the medium were determined by HPLC combined with electrochemical detection. Hypoxia augmented DA (approximately 80%) but not ACh release in naive cells, whereas, in IH-conditioned cells, it further enhanced DA release (ranging from 120 to approximately 145%) and facilitated ACh release (approximately 30%). Hypoxia-evoked augmentation of transmitter release was not seen in cells conditioned with sustained hypoxia. IH-induced increase in DA but not IH-induced ACh release during hypoxia was partially inhibited by cadmium chloride (100 microM), a voltage-gated Ca(2+) channel blocker. By contrast, 2-aminoethoxydiphenylborate (75 microM), a blocker of inositol 1,4,5-trisphosphate (IP(3)) receptors, and N-acetyl-L-cysteine (300 microM), a potent scavenger of reactive oxygen species, either attenuated or abolished IH-evoked augmentation of transmitter release during hypoxia. Together, the above results demonstrate that IH conditioning increases hypoxia-evoked neurotransmitter release from PC12 cells via mechanisms involving mobilization of Ca(2+) from intracellular stores through activation of IP(3) receptors. Our findings also suggest that oxidative stress plays a central role in IH-induced augmentation of transmitter release from PC12 cells during acute hypoxia.
Collapse
Affiliation(s)
- Dong-Kyu Kim
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106-4935, USA
| | | | | | | |
Collapse
|
40
|
Gibson GE. Interactions of oxidative stress with cellular calcium dynamics and glucose metabolism in Alzheimer's disease. Free Radic Biol Med 2002; 32:1061-70. [PMID: 12031890 DOI: 10.1016/s0891-5849(02)00802-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Considerable evidence suggests that oxidative stress (elevated levels of reactive oxygen species), altered energy metabolism, and changes in calcium dynamics are central to Alzheimer's disease (AD). Abnormalities in each of these processes occur in AD, and they can be plausibly linked to the pathology and clinical outcome of the disease. Abnormalities in these same processes in peripheral tissues, such as fibroblasts, indicate that these are inherent properties of AD cells and are not merely a secondary response to neurodegeneration. Results in cultured cells including fibroblasts demonstrate that oxidative stress can lead to the AD-related changes in calcium and energy metabolism. Data also suggest that abnormalities in the cellular calcium stores, the ability to handle oxidative stress, and to respond to metabolic impairment link the AD-causing gene mutations to the disease process. Abnormal metabolism and oxidative stress alter the proteins and cellular processes that are modified in AD, and can be readily linked to neuronal death and brain dysfunction. Prevention and/or correction of these abnormalities are appropriate therapeutic targets.
Collapse
Affiliation(s)
- Gary E Gibson
- Weill Medical College of Cornell University, Burke Medical Research Institute, White Plains, NY 10605, USA.
| |
Collapse
|